Electrophoretic methods for spatial analysis

Information

  • Patent Grant
  • 11768175
  • Patent Number
    11,768,175
  • Date Filed
    Thursday, March 4, 2021
    3 years ago
  • Date Issued
    Tuesday, September 26, 2023
    8 months ago
Abstract
The present disclosure provides electrophoretic systems, methods and compositions for spatial analysis, which can serve to magnify or demagnify spatial resolution of analytes of interest that are captured using electrophoresis. Some implementations can use a diverging or converging electric field in an electrophoretic capture system. Such a divergent or convergent electric field, as opposed to a parallel electric field, can be generated by, for example, utilizing different sizes of electrodes associated with or imbedded in substrates. Also provided herein are electrophoretic systems, methods and compositions for spatial analysis, which can serve to selectively migrate one or more analytes from a region of interest in the biological sample for capture using electrophoresis.
Description
BACKGROUND

Cells within a tissue have differences in cell morphology and/or function due to varied analyte levels (e.g., gene and/or protein expression) within the different cells. The specific position of a cell within a tissue (e.g., the cell's position relative to neighboring cells or the cell's position relative to the tissue microenvironment) can affect, e.g., the cell's morphology, differentiation, fate, viability, proliferation, behavior, signaling, and cross-talk with other cells in the tissue.


Spatial heterogeneity has been previously studied using techniques that typically provide data for a handful of analytes in the context of intact tissue or a portion of a tissue (e.g., tissue section), or provide significant analyte data from individual, single cells, but fails to provide information regarding the position of the single cells from the originating biological sample (e.g., tissue).


Various methods have been used to prepare a biological sample for analyzing analyte data in the sample. In some applications, analytes can be captured from a biological sample, and a high resolution of the captured analytes is needed for accurate analysis.


SUMMARY

The present disclosure provides electrophoretic apparatuses, systems, kits, and methods for preparing a biological sample for spatial analysis.


The spatial resolution of analytes (e.g., nucleic acids or proteins from a biological sample) may be limited due to constraints when patterning capture areas on an array designed to capture the analytes from a biological sample. The electrophoretic techniques described herein can be useful in overcoming spatial resolution limitations in a capture array system.


For example, disclosed techniques can utilize divergent or convergent electric fields, as opposed to parallel electric fields, as the basis for an electrophoretic capture array platform to enhance or improve spatial resolution of analytes. Such divergent or convergent electric fields, can be generated by different sizes of electrodes that are associated with the capture array substrates and/or the sample substrates. By way of example, a source electrode can be associated, attached to, or embedded in a sample substrate comprising a biological sample and a target electrode can be associated with a capture substrate comprising a capture probe. Alternatively, the sample substrate and the capture substrate can be made to be conductive respectively so that they can be used as anode and cathode in an electrophoretic device or system. In some instances, the source electrode can provide an effective conductive area different from the target electrode. An electric field can be generated between the source electrode and the target electrode. The ratio of the sizes of the source electrode and the target electrode can determine a degree of magnification or demagnification of the spatial resolution of analytes being captured. In some instances, the distance between the source electrode and the target electrode can be adjusted thereby allowing for the resolution to be fine tuned via magnification or demagnification. In some implementations, the distance between the electrodes can be adjusted to reduce or suppress lateral diffusion of analytes. The analytes can expand or spread out during migration from the sample substrate toward the capture substrate through the divergent electric field. The analytes captured on the capture substrate are considered to be magnified, compared to the analytes originally included in the sample on the sample substrate. The analytes contract or are concentrated while migrating from the sample substrate toward the capture substrate through the convergent electric field. The analytes captured on the capture substrate are considered to be demagnified, compared to the analytes originally included in the sample on the sample substrate.


The techniques described herein can allow for the selection of a region of interest (ROI) in a sample so that analytes in the ROI are captured via magnification or demagnification. Some implementations include an array of patterned electrodes attached to or imbedded in the source substrate (e.g., the substrate upon which a sample is located). A subset of the electrodes in the array can be selected to correspond to a ROI in the sample and actuated to generate an electric field between the selected subset of electrodes of the source substrate (e.g., comprising a biological sample) and the target substrate (e.g., the capture substrate), so that the analytes in the ROI can be captured via magnification or demagnification.


Other implementations include a light-actuated conductive film (e.g., amorphous silicon) located on the source substrate. For example, light can be illuminated onto the ROI area of the source substrate comprising a light actuated conductive film thereby generating an electrophoretic circuit between the illuminated region on the source substrate and the target substrate thereby driving analytes in the ROI from the source substrate to the target substrate where they can be captured via magnification or demagnification.


In addition, the techniques described herein provide a solution to facilitate a targeted analyte capture. For example, some implementations include a mechanism for coupling a photo-thermal effect (e.g., infrared light illumination) to thermally activate permeabilizing enzymes found in the same region as the ROI region. This can allow for the separate or sequential capture of analytes in a number of ROIs without permeabilizing the entire sample on the source substrate.


Particular embodiments described herein include an electrophoretic system for analyte migration. Such analyte migration can be used for spatial analysis of analytes in a biological sample. In some embodiments, the electrophoretic system includes a first substrate, a second substrate, a buffer chamber, and a controller. The first substrate may include a first substrate region including a biological sample. The first substrate region may be electrically conductive and have a first surface area. The second substrate may include a second substrate region for receiving analytes from the biological sample. The second substrate region may be electrically conductive and have a second surface area. The buffer chamber includes a buffer between the first substrate region and the second substrate region. The controller can generate an electric field between the first substrate region and the second substrate region such that the analytes are induced to migrate from the first substrate region (e.g., with a biological sample) toward the second substrate region (e.g., substrate for receiving analytes). The first surface area of the first substrate region may be larger or smaller than the second surface area of the second substrate region In some embodiments, first and second substrate functionalities can be reversed, such that the first substrate could receive analytes while the second substrate comprises a biological sample. In such an embodiment, an electric field is generated such that analytes are induced to migrate from the biological sample on the second substrate toward the first substrate where they are received or captured.


In some implementations, the system can optionally include one or more of the following features. The second substrate region may include capture probes for capturing the migrated analytes. In some embodiments, a ratio of the first surface area and the second surface area may range from 0.1 to 100. The first substrate region may include an array of patterned conductive regions that are selectively actuated to generate an electric field between the actuated conductive regions and the second substrate region. The first surface area of the first substrate region may be smaller than the second surface area of the second substrate region such that a generated electric field is a divergent electric field resulting in the analytes on the first substrate region being magnified on the second substrate region when captured. The first surface area of the first substrate region may be larger than the second surface area of the second substrate region such that a generated electric field is a convergent electric field resulting in the analytes on the first substrate region being demagnified on the second substrate region when captured. In some embodiments, the first substrate region is a cathode and the second substrate region is an anode.


The first substrate may be arranged in parallel with the second substrate. The first substrate may be spaced apart at a gap or distance from the second substrate. The gap or distance may be selected to maintain a diffusion rate, that is lower than a threshold diffusion rate, of the analytes migrating under an electric field. The gap or distance between a first and a second substrate may range from 1 um to 10 mm. The first substrate region and the second substrate region may have the shape of concentric circular disks, which is shaped to be circular in two dimensions. The first substrate region and the second substrate region may be shaped to be concentrically spherical in three dimensions. The first substrate and the second substrate may be glass slides and each glass slide, or regions thereof, may be coated with a conductive material. The conductive material may include at least one of tin oxide (TO), indium tin oxide (ITO), a transparent conductive oxide (TCO), aluminum doped zinc oxide (AZO), or fluorine doped tin oxide (FTO). The buffer may include a permeabilization reagent. The electrophoretic system may further include a light configured to illuminate onto at least a portion of the first substrate region to permeabilize the biological sample on the at least a portion of the first substrate region. In addition or alternatively, the sample may be permeabilized using a detergent before or after enzymatic treatment. In addition or alternatively, the sample may be incubated with a permeabilizing agent to facilitate permeabilization of the sample. In addition or alternatively, the sample may be permeabilized with a lysis reagent being added to the sample. In addition or alternatively, the sample may be permeabilized by exposing the sample to a protease capable of degrading histone proteins. The electrophoretic system may further include a power supply and electrical wires connecting the power supply to the first substrate and the second substrate, or regions thereof.


Particular embodiments described herein include a method for capturing analytes from a biological sample. The method may include placing the biological sample on a first substrate region of a first substrate, the biological sample including analytes; placing capture probes on a second substrate region of a second substrate, the second substrate region having a second surface area different from a first surface area of the first substrate region; providing a buffer between the first substrate region and the second substrate region; and generating an electric field between the first substrate region and the second substrate region to cause the analytes in the biological sample to migrate from the first substrate region toward the capture probes on the second substrate region.


In some implementations, the system can optionally include one or more of the following features. The first substrate region may include an array of patterned conductive regions. The method may include identifying a region of interest (ROI) in the biological sample, and actuating one or more of the patterned conductive regions on the first substrate to generate the electric field, the one or more of the patterned conductive regions corresponding to the ROI. The first surface area of the first substrate region may be smaller than the second surface area of the second substrate region such that the electric field is configured to be a divergent electric field, and the analytes from the ROI in the biological sample are magnified upon capture on the second substrate region. The first surface area of the first substrate region may be larger than the second surface area of the second substrate region such that the electric field is configured to be a convergent electric field, and the analytes from the ROI in the biological sample are demagnified upon capture on the second substrate region. The first substrate may be spaced apart at a distance from the second substrate. The distance may be selected to maintain a diffusion rate of the analytes migrating under the electric field. The diffusion rate is lower than a predetermined value.


Particular embodiments described herein include an electrophoretic system for capturing an analyte from a biological sample. The system may include a first substrate, a second substrate, a buffer chamber, a light generator, and a controller. The first substrate may include a first substrate region configured to place a biological sample including analytes. The biological sample may contain analytes. The first substrate region may include a photoconductive material. The second substrate may include a second substrate region configured to place capture probes thereon. The second substrate region may be configured to be conductive. The buffer chamber includes a buffer between the first substrate region and the second substrate region. The light generator may be configured to emit first light onto a least a portion of the first substrate region to permit for the at least a portion of the first substrate region to be electrically conductive. The controller may be configured to, based on the first light being emitted onto the at least a portion of the first substrate region, generate an electric field between the at least a portion of the first substrate region and the second substrate region such that the analytes in the biological sample migrate from the at least a portion of the first substrate region toward the capture probes on the second substrate region. The surface area of the at least a portion of the first substrate region may be different from a surface area of the second substrate region.


In some implementations, the system can optionally include one or more of the following features. The surface area of the at least a portion of the first substrate region may be smaller than the surface area of the second substrate region such that the electric field is configured to be a divergent electric field, and the analytes in the biological sample are magnified when captured on the second substrate region. The surface area of the at least a portion of the first substrate region is larger than the surface area of the second substrate region such that the electric field may be configured to be a convergent electric field, and the analytes in the biological sample are demagnified when captured on the second substrate region. The first substrate is a glass slide upon which is the photoconductive material including amorphous silicon. The second substrate may be a glass slide coated with a conductive material. The conductive material may include at least one of tin oxide (TO), indium tin oxide (ITO), a transparent conductive oxide (TCO), aluminum doped zinc oxide (AZO), or fluorine doped tin oxide (FTO). The first light may include at least one of an ultraviolet (UV) light, visible light, or infrared light. The electrophoretic system may include a second light configured to illuminate onto the at least a portion of the first substrate region to permeabilize the biological sample on the at least a portion of the first substrate region. The second light may be infrared light. The first substrate region may be configured to be a cathode, and the second substrate region may be configured to be an anode. The first substrate may be arranged in parallel with the second substrate. The first substrate may be arranged at an angle to the second substrate. The first substrate may be spaced apart at a distance from the second substrate. The distance may be selected to maintain a diffusion rate of the analytes migrating under the electric field to be lower than a predetermined value. The distance may range from 1 um to 10 cm. The first substrate region and the second substrate region may be shaped to be concentric circular disks. The first substrate region and the second substrate region may be shaped to be concentrically spherical. The buffer may include a permeabilization reagent. The electrophoretic system may further include a light configured to illuminate onto at least a portion of the first substrate region to permeabilize the biological sample on the at least a portion of the first substrate region. In addition or alternatively, the sample may be permeabilized using a detergent before or after enzymatic treatment. In addition or alternatively, the sample may be incubated with a permeabilizing agent to facilitate permeabilization of the sample. In addition or alternatively, the sample may be permeabilized with a lysis reagent being added to the sample. In addition or alternatively, the sample may be permeabilized by exposing the sample to a protease capable of degrading histone proteins. The electrophoretic system may include a power supply, and electrical wires connecting the power supply to the first substrate region and the second substrate region.


Particular embodiments described herein include a method for migrating analytes. The method may include placing a biological sample on a first substrate region of a first substrate, the biological sample including analytes, and the first substrate region including a photoconductive material; placing capture probes on a second substrate region of a second substrate; providing a buffer between the first substrate region and the second substrate region; emitting first light onto at least a portion of the first substrate region of the first substrate to permit for the at least a portion of the first substrate region to be electrically conductive; and generating an electric field between the at least a portion of the first substrate region and the second substrate region to cause the analytes in the biological sample to migrate from the at least a portion of the first substrate region toward the capture probes on the second substrate region. A surface area of the at least a portion of the first substrate region may be different from a surface area of the second substrate region.


In some implementations, the system can optionally include one or more of the following features. The method may further include identifying a region of interest (ROI) in the biological sample. The ROI may include the at least a portion of the first substrate region of the first substrate. The method may further include emitting second light onto the at least a portion of the first substrate region to permeabilize the biological sample on the at least a portion of the first substrate region.


All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, patent application, or item of information was specifically and individually indicated to be incorporated by reference. To the extent publications, patents, patent applications, and items of information incorporated by reference contradict the disclosure contained in the specification, the specification is intended to supersede and/or take precedence over any such contradictory material.


Where values are described in terms of ranges, it should be understood that the description includes the disclosure of all possible sub-ranges within such ranges, as well as specific numerical values that fall within such ranges irrespective of whether a specific numerical value or specific sub-range is expressly stated.


The term “each,” when used in reference to a collection of items, is intended to identify an individual item in the collection but does not necessarily refer to every item in the collection, unless expressly stated otherwise, or unless the context of the usage clearly indicates otherwise.


Various embodiments of the features of this disclosure are described herein. However, it should be understood that such embodiments are provided merely by way of example, and numerous variations, changes, and substitutions can occur to those skilled in the art without departing from the scope of this disclosure. It should also be understood that various alternatives to the specific embodiments described herein are also within the scope of this disclosure.





DESCRIPTION OF DRAWINGS

The following drawings illustrate certain embodiments of the features and advantages of this disclosure. These embodiments are not intended to limit the scope of the appended claims in any manner. Like reference symbols in the drawings indicate like elements.



FIG. 1 schematically illustrates an example electrophoretic system for preparing a sample.



FIGS. 2A-B illustrate example magnification and demagnification techniques in the electrophoretic system of FIG. 1.



FIG. 3 is a flowchart of an example process for preparing a biological sample.



FIG. 4 schematically illustrates another example electrophoretic system for preparing a sample.



FIG. 5 schematically illustrates the electrophoretic system of FIG. 4 with a mechanism for providing photo-thermal effect to permeabilize a sample.



FIG. 6 is a flowchart of an example process for preparing a biological sample.



FIG. 7 illustrates an example configuration of electrodes that can be used in the electrophoretic system.



FIG. 8 is a schematic diagram showing an example of a barcoded capture probe, as described herein.





DETAILED DESCRIPTION

In general, the techniques described herein relate to electrophoretic compositions, systems and methods for migrating analytes that may be present in a biological sample from one substrate to another. The techniques described herein can serve to magnify or demagnify spatial resolution of analytes of interest that are captured using an electrophoretic system.


Some implementations use a diverging or converging electric field in an electrophoretic capture system. Such a divergent or convergent electric field, as opposed to a parallel electric field, can be generated by utilizing different sizes of electrodes associated with or imbedded in substrates. Alternatively, a divergent or convergent electric field can also be generated by utilizing different sizes of conductive materials which may be found on substrates. Alternatively, a divergent or convergent electric field can be generated by utilizing different types of conductive materials found on substrates. In some implementations, a source electrode could be a sample substrate upon which is located a biological sample which may comprise analytes of interest, and a target electrode could be a capture substrate wherein capture probes are affixed (e.g., directly or indirectly, and reversibly or non-reversibly). The sample substrate and the capture substrate could be conductive for use as an anode and a cathode in electrophoresis. The source electrode is configured to provide an effective conductive area different from the target electrode. An electric field can be generated between the source electrode and the target electrode. The ratio of the sizes of the source electrode and the target electrode can determine a degree of magnification or demagnification of the spatial resolution of analytes being captured. Further, the distance between the source electrode and the target electrode can be adjusted thereby augmenting the degree of magnification or demagnification of the resolution. In some implementations, the distance between the electrodes can be adjusted to reduce or suppress lateral diffusion of analytes. The analytes can expand or spread out during migration from the sample substrate toward the capture substrate through the divergent electric field. The analytes captured on the capture substrate are considered to be magnified, compared to the analytes originally included in the sample on the sample substrate. The analytes contract or are concentrated while migrating from the sample substrate toward the capture substrate through the convergent electric field. The analytes captured on the capture substrate are considered to be demagnified, compared to the analytes originally included in the sample on the sample substrate


The techniques described herein allows for selecting a region of interest (ROI) in a biological sample so that the analytes in the ROI can be captured and either magnified or demagnified. Some implementations include an array of patterned electrodes from the source substrate (e.g., the substrate upon which a biological sample is located). A subset of electrodes in the source substrate can be selected that correspond to a ROI in the biological sample, and actuated to generate an electric field between the selected subset of electrodes of the source substrate and the target substrate (e.g., the capture substrate), so that the analytes in the ROI can migrate to the target substrate and be captured via magnification or demagnification as described above. For example, the selected subset of electrodes may be actuated by applying a voltage across the selected subset of electrodes to generate the electric field.


Other embodiments include a light-actuated conductive film (e.g., amorphous silicon) that can be located on the source substrate. In such an embodiment, if a light-actuated conductive film is on the source substrate, a light can be used to illuminate an ROI on the source substrate thereby generating an electrophoretic circuit between the illuminated region on the source substrate and the target substrate so that the analytes in the ROI can migrate and be captured on the target substrate. In some implementations, amorphous silicon compound can be coated on the substrate using, for example, plasma enhanced chemical vapor deposition. The photoconductive light can be on the UV and visible spectrum. Examples of amorphous silicon coating are described further in M. Ristova, et al., Study of hydrogenated amorphous silicon thin films as a potential sensor for He—Ne laser light detection, Applied Surface Science, vol. 218, Issues 1-4, 30 Sep. 2003, Pages 44-53, the disclosure of which is incorporated herein by reference in its entirety.


In addition, the techniques described herein provide a solution to facilitate a target analyte capture. For example, some implementations include a mechanism for coupling a photo-thermal effect (e.g., infrared light illumination) to thermally activate permeabilizing enzymes locally around the ROI region. This can allow for separately or sequentially capturing analytes in a number of ROIs without permeabilizing the entire sample on the source substrate. For example, water in a liquid permeabilization reagent (e.g., a buffer comprising a permeabilization agent such as a protease, e.g.,) can strongly absorb infrared light and be heated up, which is a photothermal effect. Heating can promote the enzyme activity. For example, protease enzymes (e.g., proteinase K (PROK) enzymes) can become more active at a higher temperature. In particular embodiments, the liquid permeabilization agent (e.g., the buffer comprising a permeabilization agent such as a protease, e.g., PROK) is kept at a cold temperature (e.g., around 4° C.) to suppress activity of the permeabilization agent. Local heating via, e.g., targeted infrared illumination of an ROI can locally activate the permeabilization agent such as a protease, e.g., PROK, allowing for local release of analyte from the ROI for subsequent capture. For example, the targeted infrared illumination can elevate the temperature to, e.g., 20° C.-70° C., thereby locally activating the permeabilization agent, e.g., protease such as PROK. More examples are described in Larsen, et al., Characterization of a recombinantly expressed proteinase K-like enzyme from a psychrotrophic Serratia sp., FEBS Journal, vol. 273, Issue 1, January 2006, pages 47-60 (also available at https://febs.onlinelibrary.wiley.com/doi/full/10.1111/j.1742-4658.2005.05044.x), the disclosure of which is incorporated herein by reference in its entirety.


Referring to FIGS. 1 and 2A-B, an example electrophoretic system 3000 is described. FIG. 1 schematically illustrates an example configuration of the electrophoretic system 3000. In some implementations, the electrophoretic system 3000 can be used to actively cause analytes 3014 (e.g., nucleic acids, proteins, charged molecules, etc.) in a biological sample 3012 on a source substrate 3002 to migrate to capture probes 3030 on a target substrate 3004. The electrophoretic system 3000 can improve the spatial resolution of captured analytes by actively directing analytes to diverge FIG. 2A or converge FIG. 25B toward the target substrate so that the analytes are captured on the target substrate in an expanded (e.g., magnified) or condensed (e.g., demagnified) manner. The electrophoretic system 3000 further includes an electrophoretic container 3006, a spacer 3008 between the two substrates, and a control system 3010.


Referring further to FIG. 1, the first substrate 3002 is configured to receive a biological sample 3012 that contains one or more analytes 3014. By way of example, the biological sample can be one or more cells or a tissue sample including one or more cells. The first substrate 3002 can include a first substrate 3016. In some implementations, the first substrate 3016 can include a patterned array of multiple regions, for example an array of conductive regions 3018. The biological sample 3012 can be prepared on the first substrate 3002 in various ways described herein.


The first substrate 3002 can be configured as a first electrode in the electrophoretic system 3000. For example, the first substrate 3002 can be used as a cathode. In another example, the first substrate 3002 can be used as an anode.


The first substrate 3002 can be conductive at least in the first substrate region 3016. In some implementations, the first substrate 3002 can be configured as a conductive substrate described herein. For example, the first substrate 3002 can include one or more conductive materials that permit for the first substrate 3002 to function as an electrode. Examples of such a conductive material include, but are not limited to, tin oxide (TO), indium tin oxide (ITO), a transparent conductive oxide (TCO), aluminum doped zinc oxide (AZO), fluorine doped tin oxide (FTO), and any combination thereof. Alternatively or in addition, other materials may be used to provide desired conductivity to the first substrate 3002. The first substrate 3002 can be coated with the conductive material. For example, the first substrate 3002 can include a conductive coating on the surface thereof, and the sample 3012 is provided on the coating of the substrate 3002. By way of example, the first substrate can include a glass slide coated with a conductive material.


In some embodiments, the first substrate 3002 includes an array of conductive regions 3018. The array of conductive regions 3018 can be disposed within the first substrate region 3016. Alternatively, the array of conductive regions 3018 can be disposed at least partially outside the first substrate region 3016. The conductive regions 3018 can be patterned in various configurations. In the illustrated example, the conductive regions 3018 are configured in a square pattern.


The conductive regions 3018 can be selectively activated to adjust the size of the first electrode 3020 in the electrophoretic system 3000. For example, a subset of conductive regions 3018 can be selected to correspond to a region of interest (ROI) 3022 on the sample 3012, and electrically activated to generate an electric field between the subset of conductive regions 3018 and the second substrate 3004. For example, the control system 3010 can be used to receive an input of selecting the subset of conductive regions 3018 that corresponds to the ROI 3022. In some implementations, the control system 3010 includes a user interface 3064 to receive a user input of the selection. In other implementations, the control system 3010 can automatically select the ROI 3022 or the subset of conductive regions 3018 corresponding to the ROI 3022. Further, the control system 3010 can be used to electrically activate the selected subset of conductive regions 3018 by, for example, applying a voltage across the selected subset of conductive regions 3018, thereby generating an electric field therebetween. In some implementations, the user interface 3064 can receive an input of selecting a voltage and other parameters to generate the electric field across the selected subset of conductive regions 3018.


Although the first substrate 3002 is illustrated to include a single substrate region 3016 in FIG. 1, other implementations of the first substrate 3002 can include a plurality of substrate regions configured to place multiple samples thereon, respectively. Each of such multiple substrate regions can be configured similarly to the first substrate region 3016 described herein.


Referring still to FIG. 1, the second substrate 3004 comprises capture probes 3030. In some implementations, the second substrate 3004 incudes a second substrate region 3032. The second substrate 3004 can be configured as a second electrode in the electrophoretic system 3000. For example, the second substrate 3004 can be used as an anode. In another example, the second substrate 3004 can be used as a cathode. The capture probes 3030 can be placed on the second substrate region 3032 in a variety of ways described herein. For example, capture probes 3030 can be directly attached (e.g., reversibly or non-reversibly) to a feature on an array. In another embodiment, capture probes 3030 can be indirectly attached (e.g., reversibly or non-reversibly) to a feature on an array. Alternatively or in addition, the capture probes 3030 can be immobilized on the second substrate region 3032 of the second substrate 3004. Embodiments of the capture probes 3030 are further described herein, for example with reference to FIG. 8.


The second substrate 3004 can be conductive in the second substrate region 3032. In some implementations, the second substrate 3004 can be configured as a conductive substrate described herein. For example, the second substrate 3004 can include one or more conductive materials that permit for the second substrate 3004 to function as an electrode. Examples of such a conductive material include, but are not limited to, tin oxide (TO), indium tin oxide (ITO), a transparent conductive oxide (TCO), aluminum doped zinc oxide (AZO), fluorine doped tin oxide (FTO), and any combination thereof. Alternatively or in addition, other materials may be used to provide desired conductivity to the second substrate 3004. In some implementations, the second substrate 3004 can be coated with the conductive material. For example, the second substrate 3004 can include a conductive coating on the surface thereof, and the capture probes 3030 are provided on the coating of the second substrate 3004. The second substrate can include a glass slide coated with a conductive material.


In some implementations, the second substrate 3004 can include an array of conductive regions that are selectively actuated, similarly to the conductive regions 3018 of the first substrate 3002. In some implementations, the second substrate 3004 can have a customized electrode pattern to improve capture uniformity.


In some implementations, the first substrate 3002 (or the first substrate region 3016 thereof) and the second substrate 3004 (or the second substrate region 3032 thereof) can be flat and arranged in parallel with each other. For example, the first conductive surface area of the first substrate 3002 and the second conductive surface area of the second substrate 3004 can be flat and arranged in parallel with each other. Alternatively, at least one of the first substrate 3002 (or the first substrate region 3016 thereof, or the first conductive surface area thereof) and the second substrate 3004 (or the second substrate region 3032 thereof, or the second conductive surface area thereof) can be not flat (e.g., curved or having variations in height or depth). In addition or alternatively, the first substrate 3002 (or the first substrate region 3016 thereof, or the first conductive surface area thereof) can be arranged to be angled with the second substrate 3004 (or the second substrate region 3032 thereof, or the second conductive surface area thereof). For example, in some embodiments the first substrate and the second substrate can be at a 90° angle one to the other.


In some implementations, the first substrate 3002 (or the first substrate region 3016 thereof, or the first conductive surface area thereof) and the second substrate 3004 (or the second substrate region 3032 thereof, or the second conductive surface area thereof) can be shaped as concentric circular or spherical disks. Other configurations and arrangements are also possible, such as square or rectangular shapes that are arranged concentrically or non-concentrically.


Referring to FIG. 1, the first substrate 3002 and the second substrate 3004 can be arranged within the electrophoretic container 3006. The electrophoretic container 3006 can include a buffer chamber 3050 between the first substrate 3002 and the second substrate 3004. The buffer chamber 3050 is configured to contain a buffer 3052. In some implementations, the first substrate 3002 and the second substrate 3004 can be fully immersed in the buffer 3052. In alternative implementations, either or both of the substrates 3002, 3004 can be partially immersed in the buffer 3052 contained in the electrophoretic container 3006.


The buffer 3052 can be of various types. In some implementations, the buffer 3052 includes a permeabilization reagent. In some embodiments, the buffer 3052 is contained in the buffer chamber 3050 throughout the electrophoretic process. The permeabilization reagent can permeabilize the sample before and/or during electrophoresis. In some implementations, this chemical permeabilization can be performed in conjunction with light permeabilization such as those described herein, for example with reference to FIG. 5. In addition or alternatively, the sample can be permeabilized using other methods described herein, independently or in conjunction with permeabilization using the permeabilization reagent and/or the illumination on the sample.


In some implementations, the buffer can include medium includes at least one of: a solution including a permeabilization reagent, a solid permeabilization reagent, and a hydrogel compound including a permeabilization reagent. In some embodiments, the solution including the permeabilization agent includes greater than about 2 w/v % sodium dodecyl sulfate (SDS). In some embodiments, the solution including the permeabilization agent includes about 8 w/v % to about 12 w/v % SDS. In some embodiments, the solution including the permeabilization agent includes proteinase K. In some embodiments, the solution including the permeabilization agent includes greater than 2 w/v % N-lauroylsarcosine or a sodium salt thereof. In some embodiments, the first member includes an aperture positioned so that when the first substrate is retained, the aperture is aligned with a sample region of the first substrate. In some embodiments, the second member includes at least one aperture positioned so that when the first substrate is retained and the first and second members are aligned by the alignment mechanism, anaperture of the at least one aperature is aligned with at least a portion of a sample region of the first substrate.


In some embodiments, the permeabilization reagent comprises a protease. In some embodiments, the protease is selected from trypsin, pepsin, elastase, or proteinase K.


The biological sample can be permeabilized using permeabilization reagents and techniques known in the art or otherwise described herein. Biological samples from different sources (e.g., brain, liver, ovaries, kidney, breast, colon, etc.) can require different permeabilization treatments. For example, permeabilizing the biological sample (e.g., using a protease) can facilitate the migration of analytes to the substrate surface (e.g., spatially-barcoded features). In some embodiments, the permeabilization reagents can be a detergent (e.g., saponin, Triton X100™, Tween-20™). In some embodiments, an organic solvent (e.g., methanol, acetone) can permeabilize cells of the biological sample. In some embodiments, an enzyme (e.g., trypsin) can permeabilize the biological sample. In another embodiment, an enzyme (e.g., collagenase) can permeabilize the biological sample.


In some embodiments the solution can permeabilize the biological sample and rehydrate the features (e.g., beads) of the shrunken array (e.g., shrunken hydrogel). In some embodiments, the rehydrating solution can stain the biological sample and rehydrate the features of the shrunken array (e.g., beads).


In some embodiments, the rehydrating solution (e.g., permeabilization or stain solution) can diffuse through the biological sample. In some embodiments, the rehydrating solution can reduce diffusion of analytes away from the substrate. In some embodiments, while diffusing through the biological sample, the rehydrating solution can migrate analytes toward the substrate surface and improve the efficiency of analyte capture.


Example structures and processes of using permeabilization reagents are further described in PCT Application No. PCT/US19/65100, titled Imaging System Hardware, filed Dec. 6, 2019, the disclosure of which is incorporated herein by reference in its entirety.


In some embodiments, a spacer 3008 can be disposed between the first substrate 3002 and the second substrate 3004 to separate the first substrate and the second substrate by a distance D. In some embodiments, the spacer 3008 comprises a non-conductive material, such as plastic, glass, porcelain, rubber, silicone, etc. The distance D can be determined to provide a desired level of spatial resolution based on several factors, such as, but not limited to, the strength and/or duration of an electric field generated between the first substrate 3002 and the second substrate 3004, and other parameters described herein.


The control system 3010 can generate an electric field (-E) between the first substrate 3002 and the second substrate 3004. The control system 3010 can include a controller 3060 configured to apply a voltage between the first substrate 3002 and the second substrate 3004 using a power supply 3062. The power supply 3062 can include a high voltage power supply. The controller 3060 can be electrically connected to the first substrate 3002 and the second substrate 3004, for example using electrical wires. The control system 3010 can include a user interface 3064 configured to receive a user input of starting or stopping the electrophoretic process. The user interface 3064 can include various types of input devices, such as a graphic user interface, physical or virtual buttons, switches, keypads, keyboard, etc., configured to receive a user input for adjusting operating parameters of the system 3000 or for accessing other information (e.g., instructions) associated with the system 3000. Examples of operating parameters can include, but are not limited to, voltage applied, duration of voltage application, etc. In some implementations, the input devices can be used to select a subset of conductive regions 3018 that corresponds to a region of interest (ROI) 3022 on the sample 3012 so that the subset of conductive regions are electrically activated to generate an electric field between the subset of conductive regions 3018 and the second substrate 3004. In addition, the user interface 3064 can include an output device, such as a display, lamps, etc., configured to output the operating parameters of the system 3000 or other information associated with the system 3000.


Referring still to FIG. 1, in some implementations, the first substrate 3002 has a first conductive surface area that can be used as the first electrode in the electrophoretic system 3000, and a second substrate 3004 has a second conductive surface area that can be used as the second electrode in the electrophoretic system 3000. In implementations, where the entire first substrate region 3016 is configured and used for the first electrode, the surface area of the first substrate region 3016 can be the first conductive surface area. Similarly, where the entire second substrate region 3032 is configured and used for the second electrode, the surface area of the second substrate region 3032 can be the second conductive surface area. Alternatively, where a portion of the first substrate region 3016 is configured and used for the first electrode, the surface area of the portion of the first substrate region 3016 can be the first conductive surface area. For example, where the first substrate region 3016 includes the array of conductive regions 3018, the subset of conductive regions 3018 that is activated can be the first conductive surface area. Similarly, where a portion of the second substrate region 3032 is configured and used for the second electrode, the surface area of the portion of the second substrate region 3032 can be the second conductive surface area.


In some implementations, the first conductive surface area of the first substrate 3002 is different from the second conductive surface area of the second substrate 3004, so that a divergent or convergent electric field is generated between the first and second conductive surface areas in the electrophoretic system 3000.


As illustrated in FIGS. 26 and 27A, a divergent electric field 3040 can be generated when the first conductive surface area of the first substrate 3002 is smaller than the second conductive surface area of the second substrate 3004. In the divergent electric field 3040, the analytes 3014 can migrate from the sample 3012 on the first substrate 3002 toward the second substrate 3004 in a diverging manner. For example, the analytes 3014 expand or spread out during migration through the divergent electric field 3040 prior to being captured on the second substrate 3004. In this case, the analytes 3014 captured on the second substrate 3004 are considered to be magnified, compared to the analytes originally included in the sample 3012 on the first substrate 3002.


As illustrated in FIG. 2B, a convergent electric field 3042 can be generated when the first conductive surface area of the first substrate 3002 is larger than the second conductive surface area of the second substrate 3004. In the convergent electric field 3042, the analytes 3014 can migrate from the sample 3012 on the first substrate 3002 toward the second substrate 3004 in a converging manner. For example, the analytes 3014 contract or are concentrated while migrating through the convergent electric field 3042 and captured on the second substrate 3004. In this case, the analytes 3014 captured on the second substrate 3004 are considered to be demagnified, compared to the analytes originally included in the sample 3012 on the first substrate 3002.


Magnification or demagnification can be adjusted and controlled by using different sizes of the first electrode of the first substrate 3002 and the second electrode of the second substrate 3004. For example, magnification or demagnification can be adjusting based on a ratio of the first conductive surface area of the first substrate 3002 over the second conductive surface area of the second substrate 3004. By way of example, in implementations where the first and second conductive surface areas are circular, a magnification factor M can be approximated as:






M



R

adius


of


Second


Conductive


Surface


Area


R

adius


of


First


Conductive


Surface


Area






In some implementations, the ratio between the first conductive surface area and the second conductive surface area can range between 0.1 and 100. In other implementations, the ratio between the first conductive surface area and the second conductive surface area can range between 0.01 and 1000. Other ratios are also possible.


By way of example, as illustrated in FIG. 2A, the magnification can be increased by providing a larger second conductive surface area of the second substrate 3004 than the second conductive surface area of the second substrate 3004 in FIG. 1. As illustrated in FIG. 2B, a demagnification can occur when the first conductive surface area of the first substrate 3002 is larger than the second conductive surface area of the second substrate 3004.


In addition or alternatively, the distance D between the first substrate 3002 and the second substrate 3004 can be controlled to adjust the magnification or demagnification. Further, the distance D can be controlled to suppress potential low resolution capture of the analytes that may result from lateral diffusion. For example, the distance D can be selected to maintain a diffusion rate of the analytes migrating under a divergent or convergent electric field to be lower than a predetermined value. In some implementations, the distance D can range from 1 um to 10 mm. In other implementations, the distance D can range from 0.1 um to 10 mm.



FIG. 3 is an exemplary flowchart of a process 3100 for capturing analytes from a biological sample on a sample substrate to a capture substrate using electrophoresis. In some implementations, the process 3100 can be performed using the electrophoretic system 3000 described with reference to FIGS. 1 and 2A-B. The process 3100 can include placing a sample on a first substrate (3102) and placing a capture probe on a second substrate (3104). The sample can be placed on the first substrate in various ways described herein. The capture probes can be attached to the second substrate in various ways described herein. The first and second substrates can be configured and used as electrophoretic electrodes. In some implementations, the first and second substrates can be configured as conductive substrates as described herein, such as by including a conductive material in the substrates or providing a conductive coating on an upper or lower surface of the substrates.


As described herein, a conductive surface area of the first substrate is different from a conductive surface area of the second substrate. Where the conductive surface area of the first substrate is smaller than the conductive surface area of the second substrate, a divergent electric field is generated between the first and second substrates, and the analytes can be captured on the second substrate in a magnified way. Where the conductive surface area of the first substrate is larger than the conductive surface area of the second substrate, a convergent electric field is generated between the first and second substrates, the analytes can be captured on the second substrate in a demagnified way.


The process 3100 can include providing a buffer between the first substrate and the second substrate (3106). In some implementations, the process 3100 can include arranging a spacer between the first and second substrates so that the first substrate is arranged at a distance from the second substrate. The distance can be selected to maintain a diffusion rate of the analytes migrating under the electric field to be lower than a predetermined value. As described herein, the spacer can be made of a non-conductive material and used to provide a buffer chamber between the first and second electrodes. The buffer can be contained in a buffer chamber that is provided by the spacer and used to at least partially immerse the first substrate, the second substrate, or both. In some implementations, the buffer can include a permeabilization reagent. The permeabilization reagent can permeabilize the sample before and/or during electrophoresis. In some implementations, this chemical permeabilization can be performed in conjunction with light permeabilization such as those described herein, for example with reference to FIG. 5. In addition or alternatively, the sample can be permeabilized using other methods described herein, independently or in conjunction with permeabilization using the premeabilization reagent and/or the illumination on the sample. Other buffers as described herein can be used in other implementations.


The exemplary process 3100 can include identifying a region of interest (ROI) on the first substrate (3108). The ROI can be selected to target a subset of the sample that is of particular interest and capture the analytes therefrom. The process 3100 can include generating an electric field (e.g., a divergent or convergent electric field) between the ROI of the first substrate and the second substrate (3110). For example, the first substrate can be selectively activated such that only the region of the first substrate corresponding to the ROI can be used as an electrode during electrophoresis. The electric field generated between the ROI of the first substrate and the second substrate can cause analytes to migrate from the ROI of the first substrate toward the second substrate in a diverging or converging manner depending on the type of the electric field (e.g., either a divergent electric field or a convergent electric field). In some implementations, the first substrate includes an array of patterned conductive regions that are selectively activated to correspond to the ROI of the first substrate.


Referring to FIG. 4, another exemplary electrophoretic system 3200 is described. In some implementations, the electrophoretic system 3200 can be used to actively cause analytes (e.g., nucleic acids, proteins, charged molecules, etc.) in the sample on a source substrate to migrate to capture probes on a target substrate. The electrophoretic system 3200 can improve spatial resolution of captured analytes by actively directing analytes to diverge or converge toward the target substrate so that the analytes are captured on the target substrate in an expanded (e.g., magnified) or condensed (e.g., demagnified) manner. The electrophoretic system 3200 can use a light-activated conductive material included in at least one of the source substrate and the target substrate to select a region of interest on the sample, and/or adjust a degree of magnification or demagnification of the analytes captures on the target substrate. In some implementations, the electrophoretic system 3200 includes a first substrate 3202, a second substrate 3204, an electrophoretic container 3206, a spacer 3208, and a control system 3210.


Referring still to FIG. 4, the first substrate 3202 is configured to receive a sample 3212 that contains analytes 3214. The sample 3212 includes a biological sample, such as a cell or a tissue section including a cell. The first substrate 3202 can include a first substrate region 3216 for receiving the sample 3212 thereon. The sample 3212 can be prepared on the first substrate 3202 in various ways described herein. The first substrate 3202 is configured to be used as a first electrode in the electrophoretic system 3200. For example, the first substrate 3202 can be used as a cathode. In another example, the first substrate 3202 can be used as an anode.


The first substrate 3202 can be conductive at least in the first substrate region 3216. In some implementations, the first substrate 3202 can be configured as a conductive substrate described herein. For example, the first substrate 3202 can include a light-activated conductive material 3218 that is activated to be electrically conductive when exposed to light of predetermined characteristics (e.g., wavelength). An example of such a light-activated conductive material includes amorphous silicon. See, e.g., Valley, J K et al. “Optoelectronic tweezers as a tool for parallel single-cell manipulation and stimulation.” IEEE transactions on biomedical circuits and systems vol. 3,6 (2009): 424-31. doi:10.1109/TBCAS.2009.2031329, which is hereby incorporated by reference in its entirety. Other materials may be used to provide such selective conductivity to the first substrate 3202. The light-activated conductive material 3218 can be coated to be a film or layer on the surface of the first substrate 3202 (e.g., a glass slide), and the sample 3212 is provided on the coating of the substrate 3202.


The light-activated conductive material 3218 on the first substrate 3202 can be selectively activated to adjust the size of the first electrode 3220 in the electrophoretic system 3200. The light-activated conductive material 3218 can be used to create a free form of customized region of interest (ROI) by illuminating a user-defined area on the first substrate. For example, a portion of the light-activated conductive material 3218 can be selected to correspond to a region of interest (ROI) 3222 on the sample 3212, and a beam of light 3224 is illuminated onto the portion of the light-activated conductive material 3218 so that the portion of the light-activated conductive material 3218 beomes conductive and an electric field is generated between the portion of the light-activated conductive material 3218 of the first substrate 3202 and the second substrate 3204.


Although the first substrate 3202 is illustrated to include a single substrate region 3216 in FIG. 4, other implementations of the first substrate 3202 can include a plurality of substrate regions that are configured to place multiple samples thereon, respectively. Each of such multiple substrate regions can be configured similarly to the first substrate region 3216 described herein.


Referring still to FIG. 4, the second substrate 3204 is configured to comprise capture probes 3230. In some implementations, the second substrate 3204 incudes a second substrate region 3232 configured to receive the capture probes 3230. The second substrate 3204 is configured to be used as a second electrode in the electrophoretic system 3200. For example, the second substrate 3204 can be used as an anode. In another example, the second substrate 3204 can be used as a cathode. The capture probe 3230 can be placed on the second substrate region 3232 in a variety of ways described herein. For example, the capture probes 3230 can be directly or indirectly attached to a feature that is fixed on an array. Alternatively or in addition, the capture probes 3230 can be immobilized on the second substrate region 3232 of the second substrate 3204.


The second substrate 3204 can be conductive at least in the second substrate region 3232. In some implementations, the second substrate 3204 can be configured as a conductive substrate described herein. For example, the second substrate 3204 can include one or more conductive materials that permit the second substrate 3204 to function as an electrode. Examples of such a conductive material include, but are not limited to, tin oxide (TO), indium tin oxide (ITO), a transparent conductive oxide (TCO), aluminum doped zinc oxide (AZO), fluorine doped tin oxide (FTO), and any combination thereof. Alternatively or in addition, other materials may be used to provide desired conductivity to the second substrate 3204. In some implementations, the second substrate 3204 can be coated with the conductive material. For example, the second substrate 3204 can include a conductive coating on the surface thereof, and the capture probes 3230 are provided on the coating of the second substrate 3204. The second substrate can include a glass slide coated with a conductive material.


In some implementations, the second substrate 3204 can include an array of conductive regions that are selectively actuated, similarly to the conductive regions 3018 of the first substrate 3002 described in FIG. 1. In some implementations, the second substrate 3204 can have a customized electrode pattern to improve capture uniformity. In other implementations, the second substrate 3204 can include a light-activated conductive material similarly to the first substrate 3202.


In some implementations, the first substrate 3202 (or the first substrate region 3216 thereof) and the second substrate 3204 (or the second substrate region 3232 thereof) can be flat and arranged in parallel with each other. For example, the first conductive surface area of the first substrate 3202 and the second conductive surface area of the second substrate 3204 can be flat and arranged in parallel with each other. Alternatively, at least one of the first substrate 3202 (or the first substrate region 3216 thereof, or the first conductive surface area thereof) and the second substrate 3204 (or the second substrate region 3232 thereof, or the second conductive surface area thereof) is not flat (e.g., curved). In addition or alternatively, the first substrate 3202 (or the first substrate region 3216 thereof, or the first conductive surface area thereof) can be arranged to be angled with the second substrate 3204 (or the second substrate region 3232 thereof, or the second conductive surface area thereof).


In some implementations, the first substrate 3202 (or the first substrate region 3216 thereof, or the first conductive surface area thereof) and the second substrate 3204 (or the second substrate region 3232 thereof, or the second conductive surface area thereof) can be shaped to be concentric circular or spherical disks. Other configurations and arrangements are also possible, such as square or rectangular shapes that are arranged concentrically or non-concentrically.


Referring to FIG. 4, the first substrate 3202 and the second substrate 3204 can be arranged within the electrophoretic container 3206. The electrophoretic container 3206 can provide a buffer chamber 3250 between the first substrate 3202 and the second substrate 3204. The buffer chamber 3250 is configured to contain a buffer 3252. In some implementations, the first substrate 3202 and the second substrate 3204 can be fully immersed into the buffer 3252. In alternative implementations, either or both of the substrates 3202, 3204 can be partially inserted into the buffer 3252 contained in the electrophoretic container 3206.


The buffer 3252 can be of various types. In some implementations, the buffer 3252 includes a permeabilization reagent. The buffer 3252 is contained in the buffer chamber 3250 throughout the electrophoretic process. The permeabilization reagent can permeabilize the sample before and/or during electrophoresis. In some implementations, this chemical permeabilization can be performed in conjunction with light permeabilization such as those described herein, for example with reference to FIG. 5. In addition or alternatively, the sample can be permeabilized using other methods described herein, independently or in conjunction with permeabilization using the premeabilization reagent and/or the illumination on the sample.


The spacer 3208 can be disposed between the first substrate 3202 and the second substrate 3204 to space them apart at a distance D. The spacer 3208 is made of non-conductive material, such as plastic, glass, porcelain, rubber, silicone, etc. The distance D can be determined to provide a desired level of spatial resolution based on several factors, such as the strength and/or duration of electric field generated between the first substrate 3202 and the second substrate 3204, and other parameters described herein.


The control system 3210 operates to generate an electric field (-E) between the first substrate 3202 and the second substrate 3204. The control system 3210 can include a controller 3260 configured to apply a voltage between the first substrate 3202 and the second substrate 3204 using a power supply 3262. The power supply 3262 can include a high voltage power supply. The controller 3260 can be electrically connected to the first substrate 3202 and the second substrate 3204 using electrical wires. The control system 3210 can include a user interface 3264 configured to receive a user input of starting or stopping the electrophoretic process. The user interface 3264 can include various types of input devices, such as physical or virtual buttons, GUI, switches, keypads, keyboard, etc., configured to receive a user input of adjusting operating parameters of the system 3200 or other information associated with the system 3200. Examples of such operating parameters can include a voltage being applied, a duration of such application, etc. In some implementations, the input devices can be used to select a subset of conductive regions 3218 that corresponds to a region of interest (ROI) 3222 on the sample 3212 so that the subset of conductive regions are electrically activated to generate an electric field between the subset of conductive regions 3218 and the second substrate 3204. In addition, the user interface 3264 can include an output device, such as a display, lamps, etc., configured to output the operating parameters of the system 3200 or other information associated with the system 3200.


The control system 3210 can further include a light generator 3266. The light generator 3266 is configured to generate and emit light on the first substrate 3202. Examples of light that can be used include an ultraviolet (UV) light, visible light, or infrared light. In some implementations, the light generator 3266 is manually controllable to be positioned relative to the first substrate 3202, and also manually controllable to generate and emit light to a particular area (e.g., the ROI 3222) on the first substrate 3202. For example, the light generator 3266 can be controlled based on a user input received through the user interface 3264. Alternatively or in addition, the light generator 3266 is configured to automatically adjust its position relative to the first substrate 3202 and generate and emit light toward a particular area (e.g., the ROI 3222) on the first substrate 3202.


The controller 3260 can further control the light generator 3266. In some implementations, the controller 3260 can control the light generator 3266 to arrange or orient the light generator 3266 in a desired position relative to the substrate 3202, and/or generate and project light onto the ROI 3222 so that a portion of the light-activated conductive material 3218 corresponding to the ROI 3222 is activated to be conductive.


Referring still to FIG. 4, in some implementations, the first substrate 3202 has a first conductive surface area that can be used as the first electrode in the electrophoretic system 3200, and the second substrate 3204 has a second conductive surface area that can be used as the second electrode in the electrophoretic system 3200. The first conductive surface area of the first substrate 3202 can be defined by the portion of the light-activated conductive material 3218 that is exposed to the light beam 3224. Where the entire second substrate region 3232 is configured and used for the second electrode, the surface area of the second substrate region 3232 can be the second conductive surface area. Alternatively, where a portion of the second substrate region 3232 is configured and used for the second electrode, the surface area of the portion of the second substrate region 3232 can be the second conductive surface area.


In some implementations, the first conductive surface area of the first substrate 3202 (e.g., the portion of the light-activated conductive material 3218 being exposed to light) is different from the second conductive surface area of the second substrate 3204, so that a divergent or convergent electric field is generated between the first and second conductive surface areas in the electrophoretic system 3200.


As illustrated in FIG. 4, a divergent electric field 3240 can be generated when the first conductive surface area of the first substrate 3202 (e.g., the portion of the light-activated conductive material 3218 being exposed to light) is smaller than the second conductive surface area of the second substrate 3204. In the divergent electric field 3240, the analytes 3214 can migrate from the sample 3212 on the first substrate 3202, toward the second substrate 3204, in a diverging manner. For example, the analytes 3214 are spread out during their travel through the divergent electric field 3240 and captured on the second substrate 3204. In this case, the analytes 3214 captured on the second substrate 3204 are considered to be magnified, compared to those originally included in the sample 3212 on the first substrate 3202.


In contrast, a convergent electric field can be generated when the first conductive surface area of the first substrate 3202 (e.g., the portion of the light-activated conductive material 3218 being exposed to light) is larger than the second conductive surface area of the second substrate 3204. In the convergent electric field, the analytes 3214 can migrate from the sample 3212 on the first substrate 3202, toward the second substrate 3204 in a converging manner. For example, the analytes 3214 can be concentrated while passing through the convergent electric field 3242 and captured on the second substrate 3204. In this case, the analytes 3214 captured on the second substrate 3204 are considered to be demagnified, compared to those originally included in the sample 3212 on the first substrate 3202.


Magnification or demagnification can be controlled by using different sizes of the first electrode of the first substrate 3202 (e.g., the portion of the light-activated conductive material 3218 being exposed to light) and the second electrode of the second substrate 3204. For example, magnification or demagnification can be adjusting based on a ratio of the first conductive surface area of the first substrate 3202 (e.g., the portion of the light-activated conductive material 3218 being exposed to light) over the second conductive surface area of the second substrate 3204. In some implementations, the ratio between the first conductive surface area and the second conductive surface area can range between 0.1 and 100. In other implementations, the ratio between the first conductive surface area and the second conductive surface area can range between 0.01 and 1000. Other ratios are also possible.


In addition or alternatively, the distance D between the first substrate 3202 and the second substrate 3204 can be controlled to adjust the magnification or demagnification. Further, the distance D can be controlled to suppress low resolution capture of the analytes that may result from lateral diffusion. For example, the distance D can be selected to maintain a diffusion rate of the analytes migrating under a divergent or convergent electric field to be lower than a predetermined value. In some implementations, the distance D can range from 1 um to 10 mm. In other implementations, the distance D can range from 0.1 um to 10 mm.


Referring to FIG. 5, the electrophoretic system 3200 can further include a mechanism for providing photo-thermal effect to permeabilize the sample before, during, or after operation of the electrophoretic system. For example, the control system 3210 includes a permeabilizer 3268 configured to emit a beam of light 3270 (e.g., infrared light) onto the sample placed on the first substrate 3202. In some implementations, the light 3270 can be emitted onto the same area (e.g., corresponding to the ROI 3222) to which the light beam 3224 is emitted toward the portion of the light-activated conductive material 3218. The light beam 3270 can thermally activate permeabilizing enzymes in or in proximity to the sample locally in the ROI region, thereby enhancing migration of the analytes toward the second substrate 3204.



FIG. 6 is a flowchart of an example process 3300. In some implementations, the process 3300 can be performed using the electrophoretic system 3000 described with reference to FIGS. 2 and 3. The process 3300 can include placing a sample on a first substrate (3302), and placing a capture probe on a second substrate (3304). The sample can be placed on the first substrate in various ways described herein. The capture probe can be attached to the second substrate in various ways described herein. As described herein, the first and second substrates can be configured and used as electrophoretic electrodes. In some implementations, the first and second substrates can be configured as conductive substrates as described herein, such as by including a conductive material in the substrates or providing a conductive coating on an upper or lower surface of the substrates.


As described herein, a conductive surface area of the first substrate is different from a conductive surface area of the second substrate. Where the conductive surface area of the first substrate is smaller than the conductive surface area of the second substrate, a divergent electric field is generated between the first and second substrates, and the analytes can be captured on the second substrate in a magnified way. Where the conductive surface area of the first substrate is larger than the conductive surface area of the second substrate, a convergent electric field is generated between the first and second substrates, the analytes can be captured on the second substrate in a demagnified way.


The process 3300 can include providing a buffer between the first substrate and the second substrate (3306). In some implementations, the process 3300 can include arranging a spacer between the first and second substrates so that the first substrate is arranged at a distance from the second substrate. The distance can be selected to maintain a diffusion rate of the analytes migrating under the electric field to be lower than a predetermined value. As described herein, the spacer can be made of a non-conductive material and used to provide a buffer chamber between the first and second electrodes. The buffer can be contained in a buffer chamber that is provided by the spacer and used to at least partially immerse the first substrate, the second substrate, or both. In some implementations, the buffer can include a permeabilization reagent. The permeabilization reagent can permeabilize the sample before and/or during electrophoresis. In some implementations, this chemical permeabilization can be performed in conjunction with light permeabilization such as those described herein, for example with reference to FIG. 5. In addition or alternatively, the sample can be permeabilized using other methods described herein, independently or in conjunction with permeabilization using the premeabilization reagent and/or the illumination on the sample. Other buffers as described generally herein can be used in other implementations.


The process 3300 can include identifying a region of interest (ROI) on the first substrate (3308). The ROI can be selected to target a subset of the sample that is of particular interest and capture the analytes therefrom. The process 3300 can include generating an electric field (e.g., a divergent or convergent electric field) between the ROI of the first substrate and the second substrate (3310). For example, the first substrate can be selectively activated such that only the region of the first substrate corresponding to the ROI can be used as an electrode in the electrophoresis. The electric field generated between the ROI of the first substrate and the second substrate can cause analytes to migrate from the ROI of the first substrate toward the second substrate in a diverging or converging manner depending on the type of the electric field (e.g., either a divergent electric field or a convergent electric field). In some implementations, the first substrate includes a light-activated conductive film that is activated to be electrically conductive when exposed to light of predetermined wavelength. The light-activated conductive film on the surface of the first substrate can be selectively activated to adjust the size of the first electrode in the electrophoretic system. The light-activated conductive film can be used to create a free form of customized region of interest (ROI) by illuminating a user-defined area on the first substrate.


The process 3300 can include emitting light onto the ROI of the first substrate to generate an electric field between the ROI of the first substrate and the second substrate (3310). For example, the light can be a photoconductive light that is emitted onto the ROI of the first substrate so that the region of the light-activated conductive film corresponding to the ROI is activated to be conductive. The light-emitted region (the ROI) has a size different from the second substrate so that a divergent or convergent electric field is generated between the first substrate and the second substrate.


The process 3300 can include emitting permeabilizing light onto the first substrate (3312). For example, the permeabilizing light (e.g., infrared light) can be illuminated onto the area of the sample that corresponds to the portion of the light-activated conductive film onto which the light is emitted in the step 3310. The permeabilizing light can thermally permeabilize the sample locally, thereby enhancing migration of the analytes toward the second substrate.


In some implementations, the permeabilizing light can have a wavelength close to infrared or in infrared (e.g., 1.5-2.0 um wavelengths). The photoconductive light can have a wavelength on UV or visible ranges.



FIG. 7 illustrates an example configuration of electrodes that can be used in the electrophoretic system described herein. In the illustrated example, a first electrode 3402 is a source electrode, and a second electrode 3404 is a target or capture electrode.


The first electrode 3402 can represent the first substrate 3002, 3402. Alternatively, the first electrode 3402 can represent the first substrate region 3016, 3216. Alternatively, the first electrode 3402 can represent the first conductive surface area of the first substrate 3002 described herein. For example, the first electrode 3402 can represent a portion of the conductive regions 3018 of the first substrate 3002 that is activated to be the first conductive surface area, as described herein. Alternatively, the first electrode 3402 can represent a portion of the light-activated conductive material 3218 that is light-activated to be conductive, as described herein.


The second electrode 3404 can represent the second substrate 3004, 3404. Alternatively, the second electrode 3404 can represent the second substrate region 3032, 3232. Alternatively, the second electrode 3404 can represent the second conductive surface area of the second substrate 3004 described herein.


In some implementations, as illustrated in FIG. 7, the first electrode 3402 can provide a first spherical surface 3412, and the second electrode 3404 can provide a second spherical surface 3414. The first electrode 3402 and the second electrode 3404 can be disposed concentrically so that an electric field 3420 is uniformly created without distortion between any location of the first spherical surface 3412 of the first electrode 3402 and its corresponding location of the second spherical surface 3414 of the second electrode 3404.


In other implementations, as illustrated in FIGS. 1, 2A-B, 4, and 5, the first electrode 3402 and the second electrode 3404 can be planar and arranged to be parallel with each other. The first electrode 3402 and the second electrode 3404 can be of various shapes, such as circular, rectangular, square, or triangular disks or plates. In some implementations, a potential electric field distortion that may result from the shapes of the first and second electrodes can be used to reconstruct an undistorted profile of the captured analyte.


In yet other implementations, a combination of different shapes of the first electrode 3402 and the second electrode 3404 can be used.


In some implementations, the sizes (e.g., length or diameter) of the first and second electrodes 3402, 3404 can range from 1 um to 10 mm. In other implementations, the sizes (e.g., length or diameter) of the first and second electrodes 3402, 3404 can range from 5 um to 1 cm. In some implementations, the distance between the first and second electrodes 3402, 3404 can range from 1 um to 1 cm. In other implementations, the distance between the first and second electrodes 3402, 3404 can range from 0.1 um to 10 mm.



FIG. 8 is a schematic diagram showing an exemplary capture probe, as described herein. As shown, the capture probe 102 is optionally coupled to a feature 101 by a cleavage domain 103, such as a disulfide linker. The capture probe can include a functional sequence 104 that are useful for subsequent processing. The functional sequence 104 can include all or a part of sequencer specific flow cell attachment sequence (e.g., a P5 or P7 sequence), all or a part of a sequencing primer sequence, (e.g., a R1 primer binding site, a R2 primer binding site), or combinations thereof. The capture probe can also include a spatial barcode 105. The capture probe can also include a unique molecular identifier (UMI) sequence 106. While FIG. 8 shows the spatial barcode 105 as being located upstream (5′) of UMI sequence 106, it is to be understood that capture probes wherein UMI sequence 106 is located upstream (5′) of the spatial barcode 105 is also suitable for use in any of the methods described herein. The capture probe can also include a capture domain 107 to facilitate capture of a target analyte. In some embodiments, the capture probe comprises one or more additional functional sequences that can be located, for example between the spatial barcode 105 and the UMI sequence 106, between the UMI sequence 106 and the capture domain 107, or following the capture domain 107. The capture domain can have a sequence complementary to a sequence of a nucleic acid analyte. The capture domain can have a sequence complementary to a connected probe described herein. The capture domain can have a sequence complementary to a capture handle sequence present in an analyte capture agent. The capture domain can have a sequence complementary to a splint oligonucleotide. Such splint oligonucleotide, in addition to having a sequence complementary to a capture domain of a capture probe, can have a sequence of a nucleic acid analyte, a sequence complementary to a portion of a connected probe described herein, and/or a capture handle sequence described herein.


The functional sequences can generally be selected for compatibility with any of a variety of different sequencing systems, e.g., Ion Torrent Proton or PGM, Illumina sequencing instruments, PacBio, Oxford Nanopore, etc., and the requirements thereof. In some embodiments, functional sequences can be selected for compatibility with non-commercialized sequencing systems. Examples of such sequencing systems and techniques, for which suitable functional sequences can be used, include (but are not limited to) Ion Torrent Proton or PGM sequencing, Illumina sequencing, PacBio SMRT sequencing, and Oxford Nanopore sequencing. Further, in some embodiments, functional sequences can be selected for compatibility with other sequencing systems, including non-commercialized sequencing systems.


In some embodiments, the spatial barcode 105 and functional sequences 104 is common to all of the probes attached to a given feature. In some embodiments, the UMI sequence 106 of a capture probe attached to a given feature is different from the UMI sequence of a different capture probe attached to the given feature.


Spatial analysis methodologies and compositions described herein can provide a vast amount of analyte and/or expression data for a variety of analytes within a biological sample at high spatial resolution, while retaining native spatial context. Spatial analysis methods and compositions can include, e.g., the use of a capture probe including a spatial barcode (e.g., a nucleic acid sequence that provides information as to the location or position of an analyte within a cell or a tissue sample (e.g., mammalian cell or a mammalian tissue sample) and a capture domain that is capable of binding to an analyte (e.g., a protein and/or a nucleic acid) produced by and/or present in a cell. Spatial analysis methods and compositions can also include the use of a capture probe having a capture domain that captures an intermediate agent for indirect detection of an analyte. For example, the intermediate agent can include a nucleic acid sequence (e.g., a barcode) associated with the intermediate agent. Detection of the intermediate agent is therefore indicative of the analyte in the cell or tissue sample.


Non-limiting aspects of spatial analysis methodologies and compositions are described in U.S. Pat. Nos. 10,774,374, 10,724,078, 10,480,022, 10,059,990, 10,041,949, 10,002,316, 9,879,313, 9,783,841, 9,727,810, 9,593,365, 8,951,726, 8,604,182, 7,709,198, U.S. Patent Application Publication Nos. 2020/239946, 2020/080136, 2020/0277663, 2020/024641, 2019/330617, 2019/264268, 2020/256867, 2020/224244, 2019/194709, 2019/161796, 2019/085383, 2019/055594, 2018/216161, 2018/051322, 2018/0245142, 2017/241911, 2017/089811, 2017/067096, 2017/029875, 2017/0016053, 2016/108458, 2015/000854, 2013/171621, WO 2018/091676, WO 2020/176788, Rodrigues et al., Science 363(6434):1463-1467, 2019; Lee et al., Nat. Protoc. 10(3):442-458, 2015; Trejo et al., PLoS ONE 14(2):e0212031, 2019; Chen et al., Science 348(6233):aaa6090, 2015; Gao et al., BMC Biol. 15:50, 2017; and Gupta et al., Nature Biotechnol. 36:1197-1202, 2018; the Visium Spatial Gene Expression Reagent Kits User Guide (e.g., Rev C, dated June 2020), and/or the Visium Spatial Tissue Optimization Reagent Kits User Guide (e.g., Rev C, dated July 2020), both of which are available at the 10× Genomics Support Documentation website, and can be used herein in any combination. Further non-limiting aspects of spatial analysis methodologies and compositions are described herein.


Some general terminology that may be used in this disclosure can be found in Section (I)(b) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Typically, a “barcode” is a label, or identifier, that conveys or is capable of conveying information (e.g., information about an analyte in a sample, a bead, and/or a capture probe). A barcode can be part of an analyte, or independent of an analyte. A barcode can be attached to an analyte. A particular barcode can be unique relative to other barcodes. For the purpose of this disclosure, an “analyte” can include any biological substance, structure, moiety, or component to be analyzed. The term “target” can similarly refer to an analyte of interest.


Analytes can be broadly classified into one of two groups: nucleic acid analytes, and non-nucleic acid analytes. Examples of non-nucleic acid analytes include, but are not limited to, lipids, carbohydrates, peptides, proteins, glycoproteins (N-linked or O-linked), lipoproteins, phosphoproteins, specific phosphorylated or acetylated variants of proteins, amidation variants of proteins, hydroxylation variants of proteins, methylation variants of proteins, ubiquitylation variants of proteins, sulfation variants of proteins, viral proteins (e.g., viral capsid, viral envelope, viral coat, viral accessory, viral glycoproteins, viral spike, etc.), extracellular and intracellular proteins, antibodies, and antigen binding fragments. In some embodiments, the analyte(s) can be localized to subcellular location(s), including, for example, organelles, e.g., mitochondria, Golgi apparatus, endoplasmic reticulum, chloroplasts, endocytic vesicles, exocytic vesicles, vacuoles, lysosomes, etc. In some embodiments, analyte(s) can be peptides or proteins, including without limitation antibodies and enzymes. Additional examples of analytes can be found in Section (I)(c) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. In some embodiments, an analyte can be detected indirectly, such as through detection of an intermediate agent, for example, a ligation product or an analyte capture agent (e.g., an oligonucleotide-conjugated antibody), such as those described herein.


A “biological sample” is typically obtained from the subject for analysis using any of a variety of techniques including, but not limited to, biopsy, surgery, and laser capture microscopy (LCM), and generally includes cells and/or other biological material from the subject. In some embodiments, a biological sample can be a tissue section. In some embodiments, a biological sample can be a fixed and/or stained biological sample (e.g., a fixed and/or stained tissue section). Non-limiting examples of stains include histological stains (e.g., hematoxylin and/or eosin) and immunological stains (e.g., fluorescent stains). In some embodiments, a biological sample (e.g., a fixed and/or stained biological sample) can be imaged. Biological samples are also described in Section (I)(d) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.


In some embodiments, a biological sample is permeabilized with one or more permeabilization reagents. For example, permeabilization of a biological sample can facilitate analyte capture. Exemplary permeabilization agents and conditions are described in Section (I)(d)(ii)(13) or the Exemplary Embodiments Section of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.


Array-based spatial analysis methods involve the transfer of one or more analytes from a biological sample to an array of features on a substrate, where each feature is associated with a unique spatial location on the array. Subsequent analysis of the transferred analytes includes determining the identity of the analytes and the spatial location of the analytes within the biological sample. The spatial location of an analyte within the biological sample is determined based on the feature to which the analyte is bound (e.g., directly or indirectly) on the array, and the feature's relative spatial location within the array.


A “capture probe” refers to any molecule capable of capturing (directly or indirectly) and/or labelling an analyte (e.g., an analyte of interest) in a biological sample. In some embodiments, the capture probe is a nucleic acid or a polypeptide. In some embodiments, the capture probe includes a barcode (e.g., a spatial barcode and/or a unique molecular identifier (UMI)) and a capture domain). In some embodiments, a capture probe can include a cleavage domain and/or a functional domain (e.g., a primer-binding site, such as for next-generation sequencing (NGS)). See, e.g., Section (II)(b) (e.g., subsections (i)-(vi)) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Generation of capture probes can be achieved by any appropriate method, including those described in Section (II)(d)(ii) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.


In some embodiments, more than one analyte type (e.g., nucleic acids and proteins) from a biological sample can be detected (e.g., simultaneously or sequentially) using any appropriate multiplexing technique, such as those described in Section (IV) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.


In some embodiments, detection of one or more analytes (e.g., protein analytes) can be performed using one or more analyte capture agents. As used herein, an “analyte capture agent” refers to an agent that interacts with an analyte (e.g., an analyte in a biological sample) and with a capture probe (e.g., a capture probe attached to a substrate or a feature) to identify the analyte. In some embodiments, the analyte capture agent includes: (i) an analyte binding moiety (e.g., that binds to an analyte), for example, an antibody or antigen-binding fragment thereof; (ii) analyte binding moiety barcode; and (iii) an analyte capture sequence. As used herein, the term “analyte binding moiety barcode” refers to a barcode that is associated with or otherwise identifies the analyte binding moiety. As used herein, the term “analyte capture sequence” refers to a region or moiety configured to hybridize to, bind to, couple to, or otherwise interact with a capture domain of a capture probe. In some cases, an analyte binding moiety barcode (or portion thereof) may be able to be removed (e.g., cleaved) from the analyte capture agent. Additional description of analyte capture agents can be found in Section (II)(b)(ix) of WO 2020/176788 and/or Section (II)(b)(viii) U.S. Patent Application Publication No. 2020/0277663.


There are at least two methods to associate a spatial barcode with one or more neighboring cells, such that the spatial barcode identifies the one or more cells, and/or contents of the one or more cells, as associated with a particular spatial location. One method is to promote analytes or analyte proxies (e.g., intermediate agents) out of a cell and towards a spatially-barcoded array (e.g., including spatially-barcoded capture probes). Another method is to cleave spatially-barcoded capture probes from an array and promote the spatially-barcoded capture probes towards and/or into or onto the biological sample.


In some cases, capture probes may be configured to prime, replicate, and consequently yield optionally barcoded extension products from a template (e.g., a DNA or RNA template, such as an analyte or an intermediate agent (e.g., a ligation product or an analyte capture agent), or a portion thereof), or derivatives thereof (see, e.g., Section (II)(b)(vii) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663 regarding extended capture probes). In some cases, capture probes may be configured to form ligation products with a template (e.g., a DNA or RNA template, such as an analyte or an intermediate agent, or portion thereof), thereby creating ligations products that serve as proxies for a template.


As used herein, an “extended capture probe” refers to a capture probe having additional nucleotides added to the terminus (e.g., 3′ or 5′ end) of the capture probe thereby extending the overall length of the capture probe. For example, an “extended 3′ end” indicates additional nucleotides were added to the most 3′ nucleotide of the capture probe to extend the length of the capture probe, for example, by polymerization reactions used to extend nucleic acid molecules including templated polymerization catalyzed by a polymerase (e.g., a DNA polymerase or a reverse transcriptase). In some embodiments, extending the capture probe includes adding to a 3′ end of a capture probe a nucleic acid sequence that is complementary to a nucleic acid sequence of an analyte or intermediate agent specifically bound to the capture domain of the capture probe. In some embodiments, the capture probe is extended using reverse transcription. In some embodiments, the capture probe is extended using one or more DNA polymerases. The extended capture probes include the sequence of the capture probe and the sequence of the spatial barcode of the capture probe.


In some embodiments, extended capture probes are amplified (e.g., in bulk solution or on the array) to yield quantities that are sufficient for downstream analysis, e.g., via DNA sequencing. In some embodiments, extended capture probes (e.g., DNA molecules) act as templates for an amplification reaction (e.g., a polymerase chain reaction).


Additional variants of spatial analysis methods, including in some embodiments, an imaging step, are described in Section (II)(a) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Analysis of captured analytes (and/or intermediate agents or portions thereof), for example, including sample removal, extension of capture probes, sequencing (e.g., of a cleaved extended capture probe and/or a cDNA molecule complementary to an extended capture probe), sequencing on the array (e.g., using, for example, in situ hybridization or in situ ligation approaches), temporal analysis, and/or proximity capture, is described in Section (II)(g) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Some quality control measures are described in Section (II)(h) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.


Spatial information can provide information of biological and/or medical importance. For example, the methods and compositions described herein can allow for: identification of one or more biomarkers (e.g., diagnostic, prognostic, and/or for determination of efficacy of a treatment) of a disease or disorder; identification of a candidate drug target for treatment of a disease or disorder; identification (e.g., diagnosis) of a subject as having a disease or disorder; identification of stage and/or prognosis of a disease or disorder in a subject; identification of a subject as having an increased likelihood of developing a disease or disorder; monitoring of progression of a disease or disorder in a subject; determination of efficacy of a treatment of a disease or disorder in a subject; identification of a patient subpopulation for which a treatment is effective for a disease or disorder; modification of a treatment of a subject with a disease or disorder; selection of a subject for participation in a clinical trial; and/or selection of a treatment for a subject with a disease or disorder.


Spatial information can provide information of biological importance. For example, the methods and compositions described herein can allow for: identification of transcriptome and/or proteome expression profiles (e.g., in healthy and/or diseased tissue); identification of multiple analyte types in close proximity (e.g., nearest neighbor analysis); determination of up- and/or down-regulated genes and/or proteins in diseased tissue; characterization of tumor microenvironments; characterization of tumor immune responses; characterization of cells types and their co-localization in tissue; and identification of genetic variants within tissues (e.g., based on gene and/or protein expression profiles associated with specific disease or disorder biomarkers).


Typically, for spatial array-based methods, a substrate functions as a support for direct or indirect attachment of capture probes to features of the array. A “feature” is an entity that acts as a support or repository for various molecular entities used in spatial analysis. In some embodiments, some or all of the features in an array are functionalized for analyte capture. Exemplary substrates are described in Section (II)(c) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Exemplary features and geometric attributes of an array can be found in Sections (II)(d)(i), (II)(d)(iii), and (II)(d)(iv) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.


Generally, analytes and/or intermediate agents (or portions thereof) can be captured when contacting a biological sample with a substrate including capture probes (e.g., a substrate with capture probes embedded, spotted, printed, fabricated on the substrate, or a substrate with features (e.g., beads, wells) comprising capture probes). As used herein, “contact,” “contacted,” and/or “contacting,” a biological sample with a substrate refers to any contact (e.g., direct or indirect) such that capture probes can interact (e.g., bind covalently or non-covalently (e.g., hybridize)) with analytes from the biological sample. Capture can be achieved actively (e.g., using electrophoresis) or passively (e.g., using diffusion). Analyte capture is further described in Section (II)(e) of WO 2020/176788 and/or U.S. Patent Application Publication No.


In some cases, spatial analysis can be performed by attaching and/or introducing a molecule (e.g., a peptide, a lipid, or a nucleic acid molecule) having a barcode (e.g., a spatial barcode) to a biological sample (e.g., to a cell in a biological sample). In some embodiments, a plurality of molecules (e.g., a plurality of nucleic acid molecules) having a plurality of barcodes (e.g., a plurality of spatial barcodes) are introduced to a biological sample (e.g., to a plurality of cells in a biological sample) for use in spatial analysis. In some embodiments, after attaching and/or introducing a molecule having a barcode to a biological sample, the biological sample can be physically separated (e.g., dissociated) into single cells or cell groups for analysis. Some such methods of spatial analysis are described in Section (III) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.


In some cases, spatial analysis can be performed by detecting multiple oligonucleotides that hybridize to an analyte. In some instances, for example, spatial analysis can be performed using RNA-templated ligation (RTL). Methods of RTL have been described previously. See, e.g., Credle et al., Nucleic Acids Res. 2017 Aug. 21; 45(14):e128. Typically, RTL includes hybridization of two oligonucleotides to adjacent sequences on an analyte (e.g., an RNA molecule, such as an mRNA molecule). In some instances, the oligonucleotides are DNA molecules. In some instances, one of the oligonucleotides includes at least two ribonucleic acid bases at the 3′ end and/or the other oligonucleotide includes a phosphorylated nucleotide at the 5′ end. In some instances, one of the two oligonucleotides includes a capture domain (e.g., a poly(A) sequence, a non-homopolymeric sequence). After hybridization to the analyte, a ligase (e.g., SplintR ligase) ligates the two oligonucleotides together, creating a ligation product. In some instances, the two oligonucleotides hybridize to sequences that are not adjacent to one another. For example, hybridization of the two oligonucleotides creates a gap between the hybridized oligonucleotides. In some instances, a polymerase (e.g., a DNA polymerase) can extend one of the oligonucleotides prior to ligation. After ligation, the ligation product is released from the analyte. In some instances, the ligation product is released using an endonuclease (e.g., RNAse H). The released ligation product can then be captured by capture probes (e.g., instead of direct capture of an analyte) on an array, optionally amplified, and sequenced, thus determining the location and optionally the abundance of the analyte in the biological sample.


During analysis of spatial information, sequence information for a spatial barcode associated with an analyte is obtained, and the sequence information can be used to provide information about the spatial distribution of the analyte in the biological sample. Various methods can be used to obtain the spatial information. In some embodiments, specific capture probes and the analytes they capture are associated with specific locations in an array of features on a substrate. For example, specific spatial barcodes can be associated with specific array locations prior to array fabrication, and the sequences of the spatial barcodes can be stored (e.g., in a database) along with specific array location information, so that each spatial barcode uniquely maps to a particular array location.


Alternatively, specific spatial barcodes can be deposited at predetermined locations in an array of features during fabrication such that at each location, only one type of spatial barcode is present so that spatial barcodes are uniquely associated with a single feature of the array. Where necessary, the arrays can be decoded using any of the methods described herein so that spatial barcodes are uniquely associated with array feature locations, and this mapping can be stored as described above.


When sequence information is obtained for capture probes and/or analytes during analysis of spatial information, the locations of the capture probes and/or analytes can be determined by referring to the stored information that uniquely associates each spatial barcode with an array feature location. In this manner, specific capture probes and captured analytes are associated with specific locations in the array of features. Each array feature location represents a position relative to a coordinate reference point (e.g., an array location, a fiducial marker) for the array. Accordingly, each feature location has an “address” or location in the coordinate space of the array.


Some exemplary spatial analysis workflows are described in the Exemplary Embodiments section of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. See, for example, the Exemplary embodiment starting with “In some non-limiting examples of the workflows described herein, the sample can be immersed . . . ” of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. See also, e.g., the Visium Spatial Gene Expression Reagent Kits User Guide (e.g., Rev C, dated June 2020), and/or the Visium Spatial Tissue Optimization Reagent Kits User Guide (e.g., Rev C, dated July 2020).


In some embodiments, spatial analysis can be performed using dedicated hardware and/or software, such as any of the systems described in Sections (II)(e)(ii) and/or (V) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663, or any of one or more of the devices or methods described in Sections Control Slide for Imaging, Methods of Using Control Slides and Substrates for, Systems of Using Control Slides and Substrates for Imaging, and/or Sample and Array Alignment Devices and Methods, Informational labels of WO 2020/123320.


Suitable systems for performing spatial analysis can include components such as a chamber (e.g., a flow cell or sealable, fluid-tight chamber) for containing a biological sample. The biological sample can be mounted for example, in a biological sample holder. One or more fluid chambers can be connected to the chamber and/or the sample holder via fluid conduits, and fluids can be delivered into the chamber and/or sample holder via fluidic pumps, vacuum sources, or other devices coupled to the fluid conduits that create a pressure gradient to drive fluid flow. One or more valves can also be connected to fluid conduits to regulate the flow of reagents from reservoirs to the chamber and/or sample holder.


The systems can optionally include a control unit that includes one or more electronic processors, an input interface, an output interface (such as a display), and a storage unit (e.g., a solid state storage medium such as, but not limited to, a magnetic, optical, or other solid state, persistent, writeable and/or re-writeable storage medium). The control unit can optionally be connected to one or more remote devices via a network. The control unit (and components thereof) can generally perform any of the steps and functions described herein. Where the system is connected to a remote device, the remote device (or devices) can perform any of the steps or features described herein. The systems can optionally include one or more detectors (e.g., CCD, CMOS) used to capture images. The systems can also optionally include one or more light sources (e.g., LED-based, diode-based, lasers) for illuminating a sample, a substrate with features, analytes from a biological sample captured on a substrate, and various control and calibration media.


The systems can optionally include software instructions encoded and/or implemented in one or more of tangible storage media and hardware components such as application specific integrated circuits. The software instructions, when executed by a control unit (and in particular, an electronic processor) or an integrated circuit, can cause the control unit, integrated circuit, or other component executing the software instructions to perform any of the method steps or functions described herein.


In some cases, the systems described herein can detect (e.g., register an image) the biological sample on the array. Exemplary methods to detect the biological sample on an array are described in PCT Application No. 2020/061064 and/or U.S. patent application Ser. No. 16/951,854.


Prior to transferring analytes from the biological sample to the array of features on the substrate, the biological sample can be aligned with the array. Alignment of a biological sample and an array of features including capture probes can facilitate spatial analysis, which can be used to detect differences in analyte presence and/or level within different positions in the biological sample, for example, to generate a three-dimensional map of the analyte presence and/or level. Exemplary methods to generate a two- and/or three-dimensional map of the analyte presence and/or level are described in PCT Application No. 2020/053655 and spatial analysis methods are generally described in WO 2020/061108 and/or U.S. patent application Ser. No. 16/951,864.


In some cases, a map of analyte presence and/or level can be aligned to an image of a biological sample using one or more fiducial markers, e.g., objects placed in the field of view of an imaging system which appear in the image produced, as described in the Substrate Attributes Section, Control Slide for Imaging Section of WO 2020/123320, PCT Application No. 2020/061066, and/or U.S. patent application Ser. No. 16/951,843. Fiducial markers can be used as a point of reference or measurement scale for alignment (e.g., to align a sample and an array, to align two substrates, to determine a location of a sample or array on a substrate relative to a fiducial marker) and/or for quantitative measurements of sizes and/or distances.

Claims
  • 1. A method for capturing analytes from a biological sample, the method comprising: aligning a first substrate and a second substrate, wherein the first substrate comprises the biological sample mounted on a first substrate region of the first substrate, the first substrate region including an array of selectively actuated conductive regions, the biological sample comprising the analytes, and wherein the second substrate comprises a second substrate region comprising capture probes;providing a buffer between the first substrate region and the second substrate region;selecting one or more of the selectively actuated conductive regions, such that a first surface area covered by the selected one or more of the selectively actuated conductive regions is different from a second surface area of the second substrate region; andgenerating an electric field between the selected one or more of the selectively actuated conductive regions first substrate region and the second substrate region to cause the analytes in the biological sample to migrate from the first substrate region toward the capture probes on the second substrate region.
  • 2. The method of claim 1, further comprising: emitting a first light onto at least a portion of the first substrate region of the first substrate to permit for the at least a portion of the first substrate region to be electrically conductive; andemitting a second light onto the at least a portion of the first substrate region to permeabilize the biological sample on the at least a portion of the first substrate region.
  • 3. The method of claim 2, wherein the first light is one of an ultraviolet (UV) light, visible light, or infrared light.
  • 4. The method of claim 2, wherein the second light is an infrared light.
  • 5. The method of claim 1, wherein generating the electric field comprises selectively activating the selected one or more of the selectively actuated conductive regions of the array of selectively actuated conductive regions.
  • 6. The method of claim 5, further comprising: identifying a region of interest in the biological sample, wherein the selected one or more of the selectively actuated conductive regions corresponds to the identified region of interest; andwherein the generating the electric field further comprises actuating the selected one or more of the selectively actuated conductive regions of the array of selectively actuated conductive regions.
  • 7. The method of claim 6, wherein the first surface area of the first substrate region is smaller than the second surface area of the second substrate region such that the electric field is configured to be a divergent electric field, wherein analytes from the region of interest in the biological sample are magnified upon capture on the second substrate region.
  • 8. The method of claim 6, wherein the first surface area of the first substrate region is larger than the second surface area of the second substrate region such that the electric field is configured to be a convergent electric field, wherein analytes from the region of interest in the biological sample are demagnified upon capture on the second substrate region.
  • 9. The method of claim 6, further comprising: restricting the electric field from being generated between the first substrate region and the second substrate region at other regions than the identified region of interest.
  • 10. The method of claim 9, wherein the restricting the electric field comprises: applying a voltage across only portions of the first substrate region and the second substrate region that correspond to the identified region of interest.
  • 11. The method of claim 1, wherein the first substrate is spaced apart at a distance from the second substrate, wherein the distance is selected to maintain a diffusion rate of the analytes migrating under the electric field, wherein the diffusion rate is lower than a predetermined value.
  • 12. The method of claim 11, wherein the distance ranges from 1 μm to 10 mm.
  • 13. The method of claim 1, wherein the first substrate is arranged at an angle to the second substrate.
  • 14. The method of claim 1, wherein the first substrate is arranged to be parallel with the second substrate.
  • 15. The method of claim 1, wherein the first substrate region and the second substrate region are shaped to be concentric circular disks.
  • 16. The method of claim 1, wherein the first substrate region and the second substrate region are shaped to be concentrically spherical.
  • 17. The method of claim 1, wherein the first substrate and the second substrate comprise glass slides coated with a conductive material.
  • 18. The method of claim 17, wherein the conductive material is a photoconductive material.
  • 19. The method of claim 18, wherein the photoconductive material is an amorphous silicon.
  • 20. The method of claim 18, wherein the conductive material includes at least one of tin oxide (TO), indium tin oxide (ITO), a transparent conductive oxide (TCO), aluminum doped zinc oxide (AZO), or fluorine doped tin oxide (FTO).
  • 21. The method of claim 1, wherein the capture probes each include a spatial barcode.
  • 22. The method of claim 1, wherein the capture probes includes a capture domain to facilitate capture of a target analyte from the biological sample.
  • 23. The method of claim 1, wherein the capture probes includes a unique molecular identifier (UMI) sequence.
  • 24. The method of claim 1, wherein the analytes are magnified or demagnified from the first surface area covered by the selected one or more of the selectively actuated conductive regions to the different second surface area of the second substrate region.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Patent Application Ser. No. 62/985,103 titled ELECTROPHORETIC METHODS FOR SPATIAL ANALYSIS, filed Mar. 4, 2020, the disclosure of which is hereby incorporated by reference in its entirety.

US Referenced Citations (449)
Number Name Date Kind
4683195 Mullis Jul 1987 A
4683202 Mullis Jul 1987 A
4800159 Mullis Jan 1989 A
4883867 Lee Nov 1989 A
4965188 Mullis Oct 1990 A
5002882 Lunnen Mar 1991 A
5130238 Malek Jul 1992 A
5308751 Ohkawa May 1994 A
5321130 Yue Jun 1994 A
5410030 Yue Apr 1995 A
5436134 Haugland Jul 1995 A
5455166 Walker Oct 1995 A
5494810 Barany et al. Feb 1996 A
5503980 Cantor Apr 1996 A
5512439 Hornes Apr 1996 A
5512462 Cheng Apr 1996 A
5559032 Porneroy Sep 1996 A
5582977 Yue Dec 1996 A
5599675 Brenner Feb 1997 A
5641658 Adams Jun 1997 A
5648245 Fire et al. Jul 1997 A
5658751 Yue Aug 1997 A
5716825 Hancock et al. Feb 1998 A
5750341 Macevicz May 1998 A
5763175 Brenner Jun 1998 A
5830711 Barany et al. Nov 1998 A
5837832 Chee et al. Nov 1998 A
5854033 Lizardi Dec 1998 A
5863753 Haugland Jan 1999 A
5871921 Landegren et al. Feb 1999 A
5912148 Eggerding Jun 1999 A
6013440 Lipshutz Jan 2000 A
6027889 Barany et al. Feb 2000 A
6060240 Kamb et al. May 2000 A
6083761 Kedar et al. Jul 2000 A
6130073 Eggerding Oct 2000 A
6143496 Brown Nov 2000 A
6153389 Haarer Nov 2000 A
6165714 Lane et al. Dec 2000 A
6210891 Nyren Apr 2001 B1
6210894 Brennan Apr 2001 B1
6214587 Dattagupta Apr 2001 B1
6258568 Nyren Jul 2001 B1
6266459 Walt Jul 2001 B1
6274320 Rothberg Aug 2001 B1
6300063 Lipshutz et al. Oct 2001 B1
6309824 Drmanac Oct 2001 B1
6344316 Lockhart Feb 2002 B1
6355431 Chee Mar 2002 B1
6368801 Faruqi Apr 2002 B1
6401267 Drmanac Jun 2002 B1
6404907 Gilchrist Jun 2002 B1
6432360 Church et al. Aug 2002 B1
6503713 Rana Jan 2003 B1
6506561 Cheval et al. Jan 2003 B1
6544732 Chee Apr 2003 B1
6544790 Sabatini Apr 2003 B1
6620584 Chee Sep 2003 B1
6632641 Brennan Oct 2003 B1
6673620 Loeffler Jan 2004 B1
6737236 Pieken et al. May 2004 B1
6770441 Dickinson Aug 2004 B2
6773886 Kaufman Aug 2004 B2
6787308 Balasubramanian Sep 2004 B2
6800453 Labaer Oct 2004 B2
6812005 Fan et al. Nov 2004 B2
6828100 Ronaghi Dec 2004 B1
6833246 Balasubramanian Dec 2004 B2
6859570 Walt Feb 2005 B2
6864052 Drmanac Mar 2005 B1
6897023 Fu May 2005 B2
6942968 Dickinson et al. Sep 2005 B1
7057026 Barnes Jun 2006 B2
7098041 Kaylor et al. Aug 2006 B2
7115400 Adessi Oct 2006 B1
7118883 Inoue Oct 2006 B2
7166431 Chee et al. Jan 2007 B2
7211414 Hardin May 2007 B2
7255994 Lao Aug 2007 B2
7258976 Mitsuhashi Aug 2007 B2
7297518 Quake Nov 2007 B2
7329492 Hardin Feb 2008 B2
7361488 Fan et al. Apr 2008 B2
7378242 Hurt May 2008 B2
7393665 Brenner Jul 2008 B2
7405281 Xu Jul 2008 B2
7407757 Brenner Aug 2008 B2
7537897 Brenner May 2009 B2
7563576 Chee Jul 2009 B2
7582420 Oliphant et al. Sep 2009 B2
7601498 Mao Oct 2009 B2
7635566 Brenner Dec 2009 B2
7674752 He Mar 2010 B2
7709198 Luo et al. May 2010 B2
7754429 Rigatti Jul 2010 B2
7776567 Mao Aug 2010 B2
7803943 Mao Sep 2010 B2
7910304 Drmanac Mar 2011 B2
7955794 Shen et al. Jun 2011 B2
7960119 Chee Jun 2011 B2
7960120 Rigatti Jun 2011 B2
8003354 Shen et al. Aug 2011 B2
8148068 Brenner Apr 2012 B2
8206917 Chee Jun 2012 B2
8288103 Oliphant Oct 2012 B2
8383338 Kitzman Feb 2013 B2
8460865 Chee Jun 2013 B2
8481257 Van Eijk Jul 2013 B2
8603743 Liu et al. Dec 2013 B2
8604182 Luo et al. Dec 2013 B2
8815512 Van Eijk Aug 2014 B2
8835358 Fodor Sep 2014 B2
8911945 Van Eijk Dec 2014 B2
8951726 Luo et al. Feb 2015 B2
8951781 Reed Feb 2015 B2
9062348 Van Eijk Jun 2015 B1
9194001 Brenner Nov 2015 B2
9290808 Fodor Mar 2016 B2
9290809 Fodor Mar 2016 B2
9328383 Van Eijk May 2016 B2
9334536 Van Eijk May 2016 B2
9371598 Chee Jun 2016 B2
9506061 Brown et al. Nov 2016 B2
9557330 Siciliano et al. Jan 2017 B2
9582877 Fu Feb 2017 B2
9593365 Frisen et al. Mar 2017 B2
9644204 Hindson et al. May 2017 B2
9702004 Van Eijk Jul 2017 B2
9727810 Fodor et al. Aug 2017 B2
9777324 Van Eijk Oct 2017 B2
9783841 Nolan et al. Oct 2017 B2
9834814 Peter et al. Dec 2017 B2
9850536 Oliphant et al. Dec 2017 B2
9868979 Chee et al. Jan 2018 B2
9879313 Chee et al. Jan 2018 B2
10002316 Fodor et al. Jun 2018 B2
10023907 Van Eijk Jul 2018 B2
10030261 Frisen et al. Jul 2018 B2
10041949 Bendall et al. Aug 2018 B2
10059990 Boyden et al. Aug 2018 B2
10208982 Bannish et al. Feb 2019 B2
10273541 Hindson et al. Apr 2019 B2
10472669 Chee Nov 2019 B2
10480022 Chee Nov 2019 B2
10480029 Bent et al. Nov 2019 B2
10494667 Chee Dec 2019 B2
10550429 Harada et al. Feb 2020 B2
10590244 Delaney et al. Mar 2020 B2
10724078 Van Driel et al. Jul 2020 B2
10774372 Chee et al. Sep 2020 B2
10774374 Frisen et al. Sep 2020 B2
10787701 Chee Sep 2020 B2
10913975 So et al. Feb 2021 B2
10914730 Chee et al. Feb 2021 B2
10927403 Chee et al. Feb 2021 B2
10961566 Chee Mar 2021 B2
11008607 Chee May 2021 B2
11046996 Chee et al. Jun 2021 B1
11067567 Chee Jul 2021 B2
11156603 Chee Oct 2021 B2
11162132 Frisen et al. Nov 2021 B2
11208684 Chee Dec 2021 B2
11286515 Chee et al. Mar 2022 B2
11293917 Chee Apr 2022 B2
11299774 Frisen et al. Apr 2022 B2
11313856 Chee Apr 2022 B2
11332790 Chell et al. May 2022 B2
11352659 Frisen et al. Jun 2022 B2
11359228 Chee et al. Jun 2022 B2
11365442 Chee Jun 2022 B2
11371086 Chee Jun 2022 B2
11384386 Chee Jul 2022 B2
11390912 Frisen et al. Jul 2022 B2
11401545 Chee Aug 2022 B2
11407992 Dadhwal Aug 2022 B2
11408029 Katiraee et al. Aug 2022 B2
11434524 Ramachandran Iyer et al. Sep 2022 B2
11479809 Frisen et al. Oct 2022 B2
11479810 Chee Oct 2022 B1
11492612 Dadhwal Nov 2022 B1
11505828 Chell et al. Nov 2022 B2
11512308 Gallant et al. Nov 2022 B2
11519022 Chee Dec 2022 B2
11519033 Schnall-Levin et al. Dec 2022 B2
11519138 Meier Dec 2022 B2
11535887 Gallant et al. Dec 2022 B2
11542543 Chee Jan 2023 B2
11560587 Chee Jan 2023 B2
11560592 Chew et al. Jan 2023 B2
11560593 Chell et al. Jan 2023 B2
20020040275 Cravatt Apr 2002 A1
20020150909 Stuelpnagel Oct 2002 A1
20020164611 Bamdad Nov 2002 A1
20030017451 Wang et al. Jan 2003 A1
20030022207 Balasubramanian Jan 2003 A1
20030148335 Shen et al. Aug 2003 A1
20030162216 Gold Aug 2003 A1
20030175844 Nadler Sep 2003 A1
20030215936 Kallioniemi et al. Nov 2003 A1
20030224419 Corcoran Dec 2003 A1
20030232348 Jones et al. Dec 2003 A1
20030232382 Brennan Dec 2003 A1
20040033499 Ilsley et al. Feb 2004 A1
20040067492 Peng et al. Apr 2004 A1
20040096853 Mayer May 2004 A1
20040106110 Balasubramanian Jun 2004 A1
20040112442 Maerkl Jun 2004 A1
20050037393 Gunderson et al. Feb 2005 A1
20050048580 Labaer Mar 2005 A1
20050100900 Kawashima et al. May 2005 A1
20050116161 Hafeman Jun 2005 A1
20050130173 Leamon et al. Jun 2005 A1
20050136414 Gunderson et al. Jun 2005 A1
20050179746 Roux et al. Aug 2005 A1
20050191656 Drmanac et al. Sep 2005 A1
20050191698 Chee et al. Sep 2005 A1
20050202433 Van Beuningen Sep 2005 A1
20050227271 Kwon Oct 2005 A1
20050260653 LaBaer Nov 2005 A1
20060164490 Kim et al. Jul 2006 A1
20060211001 Yu et al. Sep 2006 A1
20060216775 Burkart et al. Sep 2006 A1
20060263789 Kincaid Nov 2006 A1
20070020640 McCloskey et al. Jan 2007 A1
20070054288 Su et al. Mar 2007 A1
20070099208 Drmanac et al. May 2007 A1
20070116612 Williamson May 2007 A1
20070128624 Gormley et al. Jun 2007 A1
20070128656 Agrawal Jun 2007 A1
20070172873 Brenner et al. Jul 2007 A1
20070207482 Church et al. Sep 2007 A1
20070254305 Paik et al. Nov 2007 A1
20070269805 Hogers Nov 2007 A1
20080009420 Schroth et al. Jan 2008 A1
20080108804 Hayashizaki et al. May 2008 A1
20080132429 Perov et al. Jun 2008 A1
20080145616 Gharib et al. Jun 2008 A1
20080160580 Adessi et al. Jul 2008 A1
20080220434 Thomas Sep 2008 A1
20080261204 Lexow Oct 2008 A1
20080286795 Kawashima et al. Nov 2008 A1
20090005252 Drmanac et al. Jan 2009 A1
20090006002 Honisch et al. Jan 2009 A1
20090018024 Church et al. Jan 2009 A1
20090026082 Rothberg et al. Jan 2009 A1
20090082212 Williams Mar 2009 A1
20090099041 Church et al. Apr 2009 A1
20090105959 Braverman et al. Apr 2009 A1
20090117573 Fu et al. May 2009 A1
20090127589 Rothberg et al. May 2009 A1
20090155781 Drmanac et al. Jun 2009 A1
20090233802 Bignell et al. Sep 2009 A1
20090253581 van Eijk et al. Oct 2009 A1
20090291854 Weisinger-Mayr et al. Nov 2009 A1
20090312193 Kim et al. Dec 2009 A1
20090321262 Adachi et al. Dec 2009 A1
20100035249 Hayashizaki et al. Feb 2010 A1
20100120097 Matz et al. May 2010 A1
20100120098 Grunenwald et al. May 2010 A1
20100145037 Brive et al. Jun 2010 A1
20100159446 Haff et al. Jun 2010 A1
20100282617 Rothberg et al. Nov 2010 A1
20110028685 Purkayastha et al. Feb 2011 A1
20110059436 Hardin et al. Mar 2011 A1
20110244448 Shirai et al. Oct 2011 A1
20110245111 Chee Oct 2011 A1
20120135871 van Eijk et al. May 2012 A1
20120202698 van Eijk et al. Aug 2012 A1
20120202704 Fan et al. Aug 2012 A1
20130052331 Kram et al. Feb 2013 A1
20130096033 Routenberg Apr 2013 A1
20130171621 Luo et al. Jul 2013 A1
20140066318 Frisen et al. Mar 2014 A1
20140270435 Dunn Sep 2014 A1
20140274731 Raymond et al. Sep 2014 A1
20140323330 Glezer et al. Oct 2014 A1
20150000854 Gann-Fetter et al. Jan 2015 A1
20150148239 Jon May 2015 A1
20150219618 Krishnan Aug 2015 A1
20150292988 Bharadwaj et al. Oct 2015 A1
20160033496 Chou et al. Feb 2016 A1
20160108458 Frei et al. Apr 2016 A1
20160138091 Chee et al. May 2016 A1
20160145677 Chee et al. May 2016 A1
20160253584 Fodor et al. Sep 2016 A1
20160289740 Fu et al. Oct 2016 A1
20160298180 Chee Oct 2016 A1
20160299165 Zhou Oct 2016 A1
20170016053 Beechem et al. Jan 2017 A1
20170029875 Zhang et al. Feb 2017 A1
20170067096 Wassie et al. Mar 2017 A1
20170089811 Tillberg et al. Mar 2017 A1
20170220733 Zhuang et al. Aug 2017 A1
20170241911 Rockel et al. Aug 2017 A1
20180051322 Church et al. Feb 2018 A1
20180057873 Zhou et al. Mar 2018 A1
20180095067 Huff et al. Apr 2018 A1
20180104694 Huff et al. Apr 2018 A1
20180104964 Uemura et al. Apr 2018 A1
20180112261 Van Driel et al. Apr 2018 A1
20180201980 Chee et al. Jul 2018 A1
20180216161 Chen et al. Aug 2018 A1
20180217094 Herr et al. Aug 2018 A1
20180245142 So et al. Aug 2018 A1
20180291439 van Eijk et al. Oct 2018 A1
20180305681 Jovanovich et al. Oct 2018 A1
20190055594 Samusik et al. Feb 2019 A1
20190085383 Church et al. Mar 2019 A1
20190161796 Hauling et al. May 2019 A1
20190177777 Chee Jun 2019 A1
20190177778 Chee Jun 2019 A1
20190177789 Hindson et al. Jun 2019 A1
20190194709 Church et al. Jun 2019 A1
20190203275 Frisen et al. Jul 2019 A1
20190233878 Delaney et al. Aug 2019 A1
20190249226 Bent et al. Aug 2019 A1
20190262831 West et al. Aug 2019 A1
20190264268 Frisen et al. Aug 2019 A1
20190271030 Chee Sep 2019 A1
20190271031 Chee Sep 2019 A1
20190300943 Chee et al. Oct 2019 A1
20190300944 Chee et al. Oct 2019 A1
20190300945 Chee et al. Oct 2019 A1
20190309353 Chee Oct 2019 A1
20190309354 Chee Oct 2019 A1
20190309355 Chee Oct 2019 A1
20190323071 Chee Oct 2019 A1
20190323088 Boutet et al. Oct 2019 A1
20190330617 Church et al. Oct 2019 A1
20190338353 Belgrader et al. Nov 2019 A1
20190367969 Belhocine et al. Dec 2019 A1
20200002763 Belgrader et al. Jan 2020 A1
20200024641 Nolan et al. Jan 2020 A1
20200048690 Chee Feb 2020 A1
20200063191 Kennedy-Darling et al. Feb 2020 A1
20200063195 Chee Feb 2020 A1
20200063196 Chee Feb 2020 A1
20200071751 Daugharthy et al. Mar 2020 A1
20200080136 Zhang et al. Mar 2020 A1
20200109443 Chee Apr 2020 A1
20200224244 Nilsson et al. Jul 2020 A1
20200239946 Dewal Jul 2020 A1
20200256867 Hennek et al. Aug 2020 A1
20200277663 Iyer Sep 2020 A1
20200277664 Frenz Sep 2020 A1
20200299757 Chee et al. Sep 2020 A1
20200325531 Chee Oct 2020 A1
20200363408 Chou et al. Nov 2020 A1
20200370095 Farmer et al. Nov 2020 A1
20200399687 Frisen et al. Dec 2020 A1
20200407781 Schnall-Levin Dec 2020 A1
20210010068 Chee et al. Jan 2021 A1
20210010070 Schnall-Levin et al. Jan 2021 A1
20210095331 Fan et al. Apr 2021 A1
20210123040 Macosko et al. Apr 2021 A1
20210130881 Cox May 2021 A1
20210140982 Uytingco et al. May 2021 A1
20210150707 Weisenfeld et al. May 2021 A1
20210155982 Yin et al. May 2021 A1
20210158522 Weisenfeld et al. May 2021 A1
20210172007 Chee et al. Jun 2021 A1
20210189475 Tentori et al. Jun 2021 A1
20210190770 Delaney et al. Jun 2021 A1
20210198741 Williams Jul 2021 A1
20210199660 Williams et al. Jul 2021 A1
20210207202 Chee Jul 2021 A1
20210214785 Stoeckius Jul 2021 A1
20210222235 Chee Jul 2021 A1
20210222241 Bharadwaj Jul 2021 A1
20210222242 Ramachandran Iyer Jul 2021 A1
20210222253 Uytingco Jul 2021 A1
20210223227 Stoeckius Jul 2021 A1
20210230584 Mikkelsen et al. Jul 2021 A1
20210230681 Patterson et al. Jul 2021 A1
20210230692 Daugharthy et al. Jul 2021 A1
20210237022 Bava Aug 2021 A1
20210238664 Bava et al. Aug 2021 A1
20210238675 Bava Aug 2021 A1
20210238680 Bava Aug 2021 A1
20210247316 Bava Aug 2021 A1
20210255175 Chee et al. Aug 2021 A1
20210262018 Bava et al. Aug 2021 A1
20210262019 Alvarado Martinez et al. Aug 2021 A1
20210269864 Chee Sep 2021 A1
20210270822 Chee Sep 2021 A1
20210285036 Yin et al. Sep 2021 A1
20210285046 Chell et al. Sep 2021 A1
20210292748 Frisen et al. Sep 2021 A1
20210292822 Frisen et al. Sep 2021 A1
20210317510 Chee et al. Oct 2021 A1
20210317524 Lucero et al. Oct 2021 A1
20210324457 Ramachandran Iyer et al. Oct 2021 A1
20210332424 Schnall-Levin Oct 2021 A1
20210332425 Pfeiffer et al. Oct 2021 A1
20210348221 Chell et al. Nov 2021 A1
20220002791 Frisen et al. Jan 2022 A1
20220003755 Chee Jan 2022 A1
20220010367 Ramachandran Iyer et al. Jan 2022 A1
20220017951 Ramachandran Iyer et al. Jan 2022 A1
20220025446 Shah Jan 2022 A1
20220025447 Tentori et al. Jan 2022 A1
20220033888 Schnall-Levin et al. Feb 2022 A1
20220049293 Frenz et al. Feb 2022 A1
20220049294 Uytingco et al. Feb 2022 A1
20220064630 Bent et al. Mar 2022 A1
20220081728 Williams Mar 2022 A1
20220090058 Frisen et al. Mar 2022 A1
20220090175 Uytingco et al. Mar 2022 A1
20220090181 Gallant et al. Mar 2022 A1
20220098576 Dadhwal Mar 2022 A1
20220098661 Chew et al. Mar 2022 A1
20220106632 Galonska et al. Apr 2022 A1
20220106633 Engblom et al. Apr 2022 A1
20220112486 Ramachandran Iyer et al. Apr 2022 A1
20220112545 Chee Apr 2022 A1
20220119869 Ramachandran Iyer et al. Apr 2022 A1
20220127659 Frisen et al. Apr 2022 A1
20220127666 Katiraee et al. Apr 2022 A1
20220127672 Stoeckius Apr 2022 A1
20220145361 Frenz et al. May 2022 A1
20220154255 Chee et al. May 2022 A1
20220170083 Khaled et al. Jun 2022 A1
20220195422 Gallant et al. Jun 2022 A1
20220195505 Frisen et al. Jun 2022 A1
20220196644 Chee Jun 2022 A1
20220213526 Frisen et al. Jul 2022 A1
20220241780 Tentori et al. Aug 2022 A1
20220267844 Ramachandran Iyer et al. Aug 2022 A1
20220282329 Chell et al. Sep 2022 A1
20220290217 Frenz et al. Sep 2022 A1
20220290219 Chee Sep 2022 A1
20220298560 Frisen et al. Sep 2022 A1
20220325325 Chee et al. Oct 2022 A1
20220326251 Uytingco et al. Oct 2022 A1
20220333171 Chee Oct 2022 A1
20220333192 Uytingco Oct 2022 A1
20220333195 Schnall-Levin et al. Oct 2022 A1
20220334031 Delaney et al. Oct 2022 A1
20220348905 Dadhwal Nov 2022 A1
20220348992 Stoeckius et al. Nov 2022 A1
20220356464 Kim et al. Nov 2022 A1
20220364163 Stahl et al. Nov 2022 A1
20220389491 Chee Dec 2022 A1
20220389504 Chew et al. Dec 2022 A1
20220403455 Ramachandran Iyer et al. Dec 2022 A1
20220404245 Chell et al. Dec 2022 A1
20230002812 Stoeckius et al. Jan 2023 A1
20230014008 Shastry Jan 2023 A1
20230034039 Shahjamali Feb 2023 A1
Foreign Referenced Citations (134)
Number Date Country
3054046 Mar 2020 CA
1680604 Oct 2005 CN
1981188 Jun 2007 CN
0961110 Dec 1999 EP
0901631 Aug 2004 EP
1878502 Jan 2008 EP
1923471 May 2008 EP
2002017 Dec 2008 EP
2881465 Jun 2015 EP
3013983 May 2016 EP
3013984 May 2016 EP
2350648 Jul 2017 EP
3511423 Jul 2019 EP
3541956 Sep 2019 EP
WO 1989010977 Nov 1989 WO
WO 1991006678 May 1991 WO
WO 1995025116 Sep 1995 WO
WO 1995035505 Dec 1995 WO
WO 2002059355 Aug 2002 WO
WO 2002077283 Oct 2002 WO
WO 2003002979 Jan 2003 WO
WO 2003010176 Feb 2003 WO
WO 2005007814 Jan 2005 WO
WO-2006056861 Jun 2006 WO
WO 2007073171 Jun 2007 WO
WO 2007076726 Jul 2007 WO
WO 2007145612 Dec 2007 WO
WO 2008157801 Dec 2008 WO
WO 2009032167 Mar 2009 WO
WO 2009152928 Dec 2009 WO
WO 2010126614 Nov 2010 WO
WO 2011068088 Jun 2011 WO
WO 2011102903 Aug 2011 WO
WO 2012159089 Nov 2012 WO
WO 2013123442 Aug 2013 WO
WO 2013131962 Sep 2013 WO
WO 2013138510 Sep 2013 WO
WO 2013150082 Oct 2013 WO
WO 2013150083 Oct 2013 WO
WO 2014142841 Sep 2014 WO
WO 2014210223 Dec 2014 WO
WO 2014210225 Dec 2014 WO
WO 2016138496 Sep 2016 WO
WO 2016138500 Sep 2016 WO
WO 2016162309 Oct 2016 WO
WO 2016166128 Oct 2016 WO
WO 2016168825 Oct 2016 WO
WO 2017019456 Feb 2017 WO
WO 2017048871 Mar 2017 WO
WO 2017075293 May 2017 WO
WO 2017112957 Jun 2017 WO
WO 2017096158 Jul 2017 WO
WO 2018064640 Apr 2018 WO
WO 2018091676 May 2018 WO
WO 2018148471 Aug 2018 WO
WO 2019012005 Jan 2019 WO
WO 2019140334 Jul 2019 WO
WO 2019213254 Nov 2019 WO
WO 2019213294 Nov 2019 WO
WO 2020028194 Feb 2020 WO
WO 2020047002 Mar 2020 WO
WO 2020047004 Mar 2020 WO
WO 2020047005 Mar 2020 WO
WO 2020047010 Mar 2020 WO
WO 2020053655 Mar 2020 WO
WO 2020061064 Mar 2020 WO
WO 2020061066 Mar 2020 WO
WO 2020061108 Mar 2020 WO
WO 2020076979 Apr 2020 WO
WO 2020099640 May 2020 WO
WO 2020123301 Jun 2020 WO
WO 2020123305 Jun 2020 WO
WO 2020123309 Jun 2020 WO
WO 2020123311 Jun 2020 WO
WO 2020123316 Jun 2020 WO
WO 2020123317 Jun 2020 WO
WO 2020123318 Jun 2020 WO
WO 2020123319 Jun 2020 WO
WO 2020123320 Jul 2020 WO
WO 2020160044 Aug 2020 WO
WO 2020176788 Sep 2020 WO
WO 2020190509 Sep 2020 WO
WO 2020198071 Oct 2020 WO
WO 2020206285 Oct 2020 WO
WO 2020219901 Oct 2020 WO
WO 2020243579 Dec 2020 WO
WO 2021016379 Jan 2021 WO
WO 2021041974 Mar 2021 WO
WO 2021067246 Apr 2021 WO
WO 2021067514 Apr 2021 WO
WO 2021091611 May 2021 WO
WO 2021092433 May 2021 WO
WO 2021097255 May 2021 WO
WO 2021102003 May 2021 WO
WO 2021102005 May 2021 WO
WO 2021102039 May 2021 WO
WO 2021133845 Jul 2021 WO
WO 2021133849 Jul 2021 WO
WO 2021142233 Jul 2021 WO
WO 2021168261 Aug 2021 WO
WO 2021168278 Aug 2021 WO
WO 2021216708 Oct 2021 WO
WO 2021225900 Nov 2021 WO
WO 2021236625 Nov 2021 WO
WO 2021236929 Nov 2021 WO
WO 2021237056 Nov 2021 WO
WO 2021237087 Nov 2021 WO
WO 2021242834 Dec 2021 WO
WO 2021247543 Dec 2021 WO
WO 2021247568 Dec 2021 WO
WO 2021252499 Dec 2021 WO
WO 2021252576 Dec 2021 WO
WO 2021252591 Dec 2021 WO
WO 2021252747 Dec 2021 WO
WO 2021263111 Dec 2021 WO
WO 2022025965 Feb 2022 WO
WO 2022060798 Mar 2022 WO
WO 2022060953 Mar 2022 WO
WO 2022061152 Mar 2022 WO
WO 2022087273 Apr 2022 WO
WO 2022099037 May 2022 WO
WO 2022109181 May 2022 WO
WO 2022140028 Jun 2022 WO
WO 2022147005 Jul 2022 WO
WO 2022147296 Jul 2022 WO
WO 2022164615 Aug 2022 WO
WO 2022178267 Aug 2022 WO
WO 2022198068 Sep 2022 WO
WO 2022221425 Oct 2022 WO
WO 2022226057 Oct 2022 WO
WO 2022236054 Nov 2022 WO
WO 2022256503 Dec 2022 WO
WO 2022271820 Dec 2022 WO
WO 2023287765 Jan 2023 WO
Non-Patent Literature Citations (181)
Entry
K.A. Carter, Porphyrin-phospholipid liposomes premeablizied by near-infrared light, Nat. Commun., 2014, 5:3546, p. 1-11. (Year: 2014).
U.S. Appl. No. 16/353,937, filed Mar. 14, 2019, Frisen et al.
U.S. Appl. No. 17/707,189, filed Mar. 29, 2022, Chell et al.
Borm et al., “Scalable in situ single-cell profiling by electrophoretic capture of mRNA,” bioRxiv, Jan. 2022, 32 pages.
Chen et al., “Gray-scale photolithography using microfluidic photomasks,” PNAS, Feb. 2003, 100(4):1499-1504.
Dalma-Weiszhausz et al., “The affymetrix GeneChip platform: an overview,” Methods Enzymol., 2006, 410:3-28.
Miller et al., “Chapter 11—Solid and Suspension Microarrays for Microbial Diagnostics,” Methods in Microbiology, 2015, 42:395-431.
Vickovic et al., “SM-Omics: An automated Platform for High-Throughput Spatial Multi-Omics,” bioRxiv, Oct. 2020, 40 pages.
assets.ctassets.net [online], “Technical Note—Visium Spatial Gene Expression Imaging Guidelines,” CG000241 Rev A, 2019, retrieved on Jul. 29, 2022, retrieved from URL <https://assets.ctfassets.net/an68im79xiti/76JHgFQo6aLq8UPyfL0u2c/fc39e46f86bf75676d3f7da6dc721fad/CG000241_VisiumImagingGuidelinesTN_Rev_A.pdf>, 8 pages.
Burgess, “Spatial transcriptomics coming of age,” Nat Rev Genet., Jun. 2019, 20(6):317, 1 page.
Cardona et al., “TrakEM2 0.9a User Manual,” Sep. 8, 2011, retrieved on Jul. 29, 2022, retreieved from URL <https://www.ini.uzh.ch/˜acardona/trakem2_manual.html>, 38 pages.
github.com [online], “ST Spot Detector Usage Guide: A Guide to Using the Spatial Transcriptomics Spot Detector 2.0,” Jun. 2018, retrieved on Jul. 29, 2022, retrieved from URL <https://github.com/SpatialTranscriptomicsResearch/st_spot_detector/wiki/ST-Spot-Detector-Usage-Guide, 6 pages.
Navarro et al., “ST viewer: a tool for analysis and visualization of spatial transcriptomics datasets: Supplementary Information,” Bioinformatics, Mar. 2019, 1058-1060.
Wilbrey-Clark et al., “Cell Atlas technologies and insights into tissue architecture,” Biochemical Journal, Apr. 2020, 477(8):1427-1442.
Wong et al., “ST Spot Detector: a web-based application for automatic spot and tissue detection for spatial Transcriptomics image datasets,” Bioinformatics, Jan. 2018, 34(11):1966-1968.
Asp et al., “Spatially Resolved Transcriptomes—Next Generation Tools for Tissue Exploration,” Bioessays, Oct. 2020, 42(10):e1900221, 16 pages.
Bergenstråhle et al., “Seamless integration of image and molecular analysis for spatial transcriptomics workflows,” BMC Genomics, Jul. 2020, 21(1):482, 7 pages.
Bolotin et al., “MiXCR: software tor comprehensive adaptive immunity profiling,” Nat Methods., May 2015, 12(5):380-1.
U.S. Appl. No. 63/033,348, filed Jun. 2, 2020, Bent.
Borm et al., “High throughput Human embryo spatial transcriptome mapping by surface transfer of tissue RNA,” Abstracts Selected Talks, Single Cell Genomics mtg, (SCG2019), 2019, 1 pages (Abstract Only).
Chen et al., “Large field of view-spatially resolved transcriptomics at nanoscale resolution,” bioRxiv, Jan. 19, 2021, retrieved from URL <https://www.biorxiv.org/node/1751045.abstract>, 37 pages.
Cho et al., “Seq-Scope: Submicrometer-resolution spatial transcriptomics for single cell and subcellular studies,” bioRxiv, Jan. 27, 2021, retrieved from URL <https://www.biorxiv.org/node/1754517.abstract>, 50 pages.
Codeluppi et al., “Spatial organization of the somatosensory cortex revealed by osmFISH,” Nature Methods, Nov. 2018, 15:932-935.
Eng et al., “Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH+,” Nature, Apr. 2019, 568(7751):235-239, 37 pages.
Gamper et al., “Gene expression profile of bladder tissue of patients with ulcerative interstitial cystitis,” BMC Genomics, Apr. 28, 2009, 10(199):1-17.
Goh et al., “Highly Specific Multiplexed RNA Imaging in Tissues With Split-FISH,” Nat Methods, Jun. 15, 2020, 17(7):689-693, 21 pages.
Liu et al., “High-Spatial-Resolution Multi-Omnics Sequencing via Deterministic Barcoding in Tissue,” Cell, Nov. 13, 2020, 183(6):1665-1681, 36 pages.
Liu et al., “Spatial transcriptome sequencing of FFPE tissues at cellular level,” bioRxiv 788992, Oct. 14, 2020, 39 pages.
Takei et al., “Integrated Spatial Genomics Reveals Global Architecture of Single Nuclei,” Nature, Jan. 27, 2021, 590(7845):344-350, 53 pages.
Xia et al., “Spatial transcriptome profiling by MERFISH reveals subcellular RNA compartmentalization and cell cycle-dependent gene expression”, Proceedings of the National Academy of Sciences, Sep. 2019, 116(39):19490-19499.
U.S. Appl. No. 16/876,709, filed May 18, 2020, Schnall-Levin et al.
U.S. Appl. No. 61/839,313, filed Jun. 25, 2013, Chee et al.
U.S. Appl. No. 61/839,320, filed Jun. 25, 2013, Chee et al.
[No Author Listed], “ChromiumNext GEM Single Cell 3′ Reagent Kits v3.1—User Guide,” 10x Genomics, Document No. CG000204, Nov. 2019, 58 pages.
[No Author Listed], “Chromium Next GEM Single Cell 3′ Reagent Kits v3.1 (Dual Index)—User Guide,” 10x Genomics, Mar. 2021, Document No. CG000315, 61 pages.
[No Author Listed], “HuSNP Mapping Assay User's Manual,” Affymetrix Part No. 90094 (Affymetrix, Santa Clara, Calif.), GeneChip, 2000, 104 pages.
[No Author Listed], “Microarray technologies have excellent possibilities in genomics-related researches,” Science Tools From Amersham Pharmacia Biotech, 1998, 3(4): 8 pages (with English Translation).
10xGenomics.com, [online], “Visium Spatial Gene Expression Reagent Kits—Tissue Optimization—User Guide,” Jul. 2020, retrieved on May 25, 2021, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/5UJrN0cH17rEk0UXwd19It/e54d99fb08a8f1500aba503005a04a56/CG000238_VisiumSpatialTissueOptimizationUserGuide_RevD.pdf>, 42 pages.
10xGenomics.com, [online], “Visium Spatial Gene Expression Reagent Kits—Tissue Optimization,” Nov. 2019, retrieved on Jan. 25, 2022, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/4q03w6959AJFxffSw51ee9/6a2ac61cf6388a72564eeb96bc294967/CG000238_VisiumSpatialTissueOptimizationUserGuide_Rev_A.pdf>, 46 pages.
10xGenomics.com, [online], “Visium Spatial Gene Expression Reagent Kits—Tissue Optimization,” Oct. 2020, retrieved on Dec. 28, 2021, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/5UJrN0cH17rEk0UXwd19It/e54d99fb08a8f1500aba503005a04a56/CG000238_VisiumSpatialTissueOptimizationUserGuide_RevD.pdf>, 43 pages.
10xGenomics.com, [online], “Visium Spatial Gene Expression Reagent Kits—User Guide,” Jun. 2020, retrieved on May 25, 2021, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/3GGIfH3RWpd1bFVhalpexR/8baa08d9007157592b65b2cdc7130990/CG000239_VisiumSpatialGeneExpression_UserGuide_RevD.pdf>, 69 pages.
10xGenomics.com, [online], “Visium Spatial Gene Expression Reagent Kits—User Guide,” Oct. 2020, retrieved on Dec. 28, 2021, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/3GGIfH3RWpd1bFVhalpexR/8baa08d9007157592b65b2cdc7130990/CG000239_VisiumSpatialGeneExpression_UserGuide_RevD.pdf>, 70 pages.
Adessi et al., “Solid phase DNA amplification: characterisation of primer attachment and amplification mechanisms,” Nucl. Acids Res., 2000, 28(20):E87, 8 pages.
Affymetrix, “GeneChip Human Genome U133 Set,” retrieved from the Internet: on the World Wide Web at affymetrix.com/support/technical/datasheets/hgu133_datasheet.pdf, retrieved on Feb. 26, 2003, 2 pages.
Affymetrix, “Human Genome U95Av2,” Internet Citation, retrieved from the internet: on the World Wide Web affymetrix.com, retrieved on Oct. 2, 2002, 1 page.
Albretsen et al., “Applications of magnetic beads with covalently attached oligonucleotides in hybridization: Isolation and detection of specific measles virus mRNA from a crude cell lysate,” Anal. Biochem., 1990, 189(1):40-50.
Allawi et al., “Thermodynamics and NMR of Internal GâT Mismatches in DNA,” Biochemistry, 1996, 36(34):10581-10594.
Angenendt et al., “Cell-free Protein expression and functional assay in a nanowell chip format,” Analytical Chemistry, 2004, 76(7):1844-49.
Angenendt et al., “Generation of High Density Protein Microarrays by Cell-free in Situ Expression of Unpurified PCR Products,” Molecular and Cellular Proteomics, (2006) Ch. 5.9, pp. 1658-1666.
Armani et al, “2D-PCR: a method of mapping DNA in tissue sections,” Lab Chip, 2009, 9(24):3526-34.
Atkinson et al., “An Updated Protocol for High Throughput Plant Tissue Sectioning,” Front Plant Sci, 2017, 8:1721, 8 pages.
Atkinson, “Overview of Translation: Lecture Manuscript,” U of Texas, 2000, DD, pp. 6.1-6.8.
Bains et al., “A novel method for nucleic acid sequence determination,” Journal of Theoretical Biology, 1988, 135(3), 303-7.
Barnes, “PCR amplification of up to 35-kb DNA with high fidelity and high yield from lambda bacteriophage templates,” Proc. Natl. Acad. Sci USA, 1994, 91(6):2216-2220.
Baugh et al., “Quantitative analysis of mRNA amplification by in vitro transcription,” Nucleic Acids Res., 2001, 29(5):e29, 9 pages.
Beattie et al., “Advances in genosensor research,” Clin Chem., May 1995, 41(5):700-6.
Beechem et al., “High-Plex Spatially Resolved RNA and Protein Detection Using Digital Spatial Profiling: A Technology Designed for Immuno-oncology Biomarker Discovery and Translational Research,” Methods Mol Biol, 2020, Chapter 25, 2055:563-583.
Birney et al., “Identification and analysis of functional elements in 1% of the human genome by the ENCODE pilot project,” Nature, 2007, 447(7146):799-816.
Blanchard et al., “High-density oligonucleotide arrays,” Biosensors & Bioelectronics, 1996, 11(6-7):687-690.
Blokzijl et al., “Profiling protein expression and interactions: proximity ligation as a tool for personalized medicine,” J Intern. Med., 2010, 268(3):232-245.
Blow, “Tissue Issues,” Nature, 2007, 448(7156):959-962.
Brandon et al., “Mitochondrial mutations in cancer,” Oncogene, 2006, 25(34):4647-4662.
Brenner et al., “Gene expression analysis by massively parallel signature sequencing (MPSS) on microbead arrays,” Nat. Biotech., 2000, 18(6):630-634.
Brenner et al., “In vitro cloning of complex mixtures of DNA on microbeads: physical separation of differentially expressed cDNAs,” Proc. Natl. Acad. Sci. USA, 2000, 97(4):1665-1670.
Brow, “35—The Cleavase I enzyme for mutation and polymorphism scanning,” PCR Applications Protocols for Functional Genomics, 1999, pp. 537-550.
Brown et al., “Retroviral integration: structure of the initial covalent product and its precursor, and a role for the viral IN protein,” Proc Natl Acad Sci USA, Apr. 1989, 86(8):2525-9.
Buenrostro et al., “Transposition of native chromatin for multimodal regulatory analysis and personal epigenomics,” Nat Methods, Dec. 2013, 10(12):1213-1218.
Bullard et al., “Direct comparison of nick-joining activity of the nucleic acid ligases from bacteriophage T4,” Biochem. J. 2006, 398(1):135-144.
Burgess, “A space for transcriptomics,” Nature Reviews Genetics, 2016, 17(8):436-7.
Burgess, “Finding structure in gene expression,” Nature Reviews Genetics, 2018, 19(5):249, 1 page.
Burns et al., “Well-less, gel-permeation formats for ultra-HTS,” DDT, 2001, 6(12):S40-S47.
Burton et al., “Coverslip Mounted-Immersion Cycled in Situ RT-PCR for the Localization of mRNA in Tissue Sections,” Biotechniques, 1998, 24(1):92-100.
Cha et al., “Specificity, efficiency, and fidelity of PCR,” Genome Res., 1993, 3(3):S18-29.
Chandra et al., “Cell-free synthesis-based protein microarrays and their applications,” Proteomics, 2009, 5(6):717-30.
Chatterjee et al., “Mitochondrial DNA mutations in human cancer. Oncogene,” 2006, 25(34):4663-4674.
Chen et al., “RNA imaging. Spatially resolved, highly multiplexed RNA profiling in single cells,” Science, Apr. 2015, 348(6233):aaa6090, 21 pages.
Chen et al., “Spatial Transcriptomics and In Situ Sequencing to Study Alzheimer's Disease,” Cell, Aug. 2020, 182(4):976-991.
Chen et al., “μCB-seq: microfluidic cell barcoding and sequencing for high-resolution imaging and sequencing of single cells,” Lab Chip, Nov. 2020, 20(21):3899-3913.
Constantine et al., “Use of genechip high-density oligonucleotide arrays for gene expression monitoring,” Life Sceience News, Amersham Life Science, 1998, pp. 11-14.
Credle et al., “Multiplexed analysis of fixed tissue RNA using Ligation in situ Hybridization,” Nucleic Acids Research, 2017, 45(14):e128, 9 pages.
Crosetto et al., “Spatially resolved transcriptomics and beyond,” Nature Review Genetics, 2015, 16(1):57-66.
Czarnik, “Encoding methods for combinatorial chemistry,” Curr Opin Chem Biol., Jun. 1997, 1(1):60-6.
Dahl et al., “Circle-to-circle amplification for precise and sensitive DNA analysis,” Proc. Natl. Acad. Sci., 2004, 101(13):4548-4553.
Daubendiek et al., “Rolling-Circle RNA Synthesis: Circular Oligonucleotides as Efficient Substrates for T7 RNA Polymerase,” J. Am. Chem. Soc., 1995, 117(29):7818-7819.
Davies et al., “How best to identify chromosomal interactions: a comparison of approaches,” Nat. Methods, 2017, 14(2):125-134.
Dean et al., “Comprehensive human genome amplification using multiple displacement amplification,” Proc Natl. Acad. Sci. USA, 2002, 99(8):5261-66.
Duncan et al., “Affinity chromatography of a sequence-specific DNA binding protein using Teflon-linked oligonucleotides,” Anal. Biochem., 1988, 169(1):104-108.
Eguiluz et al., “Multitissue array review: a chronological description of tissue array techniques, applications and procedures,” Pathology Research and Practice, 2006, 202(8):561-568.
Eldridge et al., “An in vitro selection strategy for conferring protease resistance to ligand binding peptides,” Protein Eng Des Sel., 2009, 22(11):691-698.
Ellington et al., “Antibody-based protein multiplex platforms: technical and operational challenges,” Clin Chem, 2010, 56(2):186-193.
Fahy et al., “Design and synthesis of polyacrylamide-based oligonucleotide supports for use in nucleic acid diagnostics,” Nucleic Acids Res., Apr. 1993, 21(8):1819-26.
Fan et al., “Highly parallel SNP genotyping,” Cold Spring Symp. Quant. Biol., 68: 69-78, 2003.
Fire et al., “Rolling replication of short DNA circles,” Proc. Natl. Acad. Sci., 1995, 92(10):4641-4645.
Fodor et al., “Light-directed, spatially addressable parallel chemical synthesis,” Science, 1995, 251(4995):767-773.
Forster et al., “A human gut bacterial genome and culture collection for improved metagenomic analyses,” Nature Biotechnology, 2019, 37(2):186-192.
Frese et al., “Formylglycine aldehyde Tag—protein engineering through a novel post-translational modification,” ChemBioChem., 2009, 10(3):425-27.
Fu et al., “Counting individual DNA molecules by the stochastic attachment of diverse labels,” PNAS, 2011, 108(22):9026-9031.
Fu et al., “Continuous Polony Gels for Tissue Mapping with High Resolution and RNA Capture Efficiency,” bioRxiv, 2021, 20 pages.
Fullwood et al., “Next-generation DNA sequencing of paired-end tags (PET) for transcriptome and genome analyses,” Genome Res., 2009, 19(4):521-532.
Ganguli et al., “Pixelated spatial gene expression analysis from tissue,” Nat Commun., Jan. 2018, 9(1):202, 9 pages.
Gao et al., “High density peptide microarrays. In situ synthesis and applications,” Molecular Diversity, 8, 177-187, 2004.
Gao et al., “Q&A: Expansion microscopy”, BMC Biology, 15:50, 9 pages, 2017.
Gene@arrays[online], BeadArray Technology, available on or before Feb. 14, 2015, via Internet Archive: Wayback Machine URL <https://web.archive.org/web/20150214084616/http://genearrays.com/services/microarrays/illumina/beadarray-technology/>, [retrieved on Jan. 30, 2020], 3 pages.
Gnanapragasam, “Unlocking the molecular archive: the emerging use of formalin-fixed paraffin-embedded tissue for biomarker research in urological cancer,” BJU International, 2009, 105(2):274-278.
Goldkorn et al., “A simple and efficient enzymatic method for covalent attachment of DNA to cellulose. Application for hybridization-restriction analysis and for in vitro synthesis of DNA probes,” Nucleic Acids Res., 1986, 14(22):9171-9191.
Gracia Villacampa et al., “Genome-wide Spatial Expression Profiling in FFPE Tissues,” bioRxiv, 2020, pp. 38 pages.
Gunderson et al., “Decoding randomly ordered DNA arrays,” Genome Research, 2004, 14(5):870-877.
Guo et al., “Direct fluorescence analysis of genetic polymorphisms by hybridization with oligonucleotide arrays on glass supports,” Nucleic Acids Res., Dec. 1994, 22(24):5456-65.
Gupta et al., “Single-cell isoform RNA sequencing characterizes isoforms in thousands of cerebellar cells,” Nature Biotechnol., Oct. 2018, 36:1197-1202.
Hamaguchi et al., “Direct reverse transcription-PCR on oligo(dT)-immobilized polypropylene microplates after capturing total mRNA from crude cell lysates,” Clin Chem., Nov. 1998, 44(11):2256-63.
Hayes et al., “Electrophoresis of proteins and nucleic acids: I-Theory,” BMJ, Sep. 1989, 299(6703):843-6.
He et al., “In situ synthesis of protein arrays,” Current Opinion in Biotechnology, 2008, 19(1):4-9.
He, “Cell-free protein synthesis: applications in proteomics and biotechnology,” New Biotechnology, 2008, 25(2-3):126-132.
Hejatko et al., “In situ hybridization technique for mRNA detection in whole mount Arabidopsis samples,” Nature Protocols, 2006, 1(4):1939-1946.
Hiatt et al., “Parallel, tag-directed assembly of locally derived short sequence reads,” Nature Methods, 2010, 7(2):119-25.
Jamur et al., “Permeabilization of cell membranes.,” Method Mol. Biol., 2010, 588:63-66.
Jemt et al., “An automated approach to prepare tissue-derived spatially barcoded RNA-sequencing libraries,” Scientific Reports, 2016, 6:37137, 10 pages.
Kapteyn et al., “Incorporation of non-natural nucleotides into template-switching oligonucleotides reduces background and improves cDNA synthesis from very small RNA samples,” BMC Genomics, Jul. 2010, 11:413, 9 pages.
Korbel et al., “Paired-end mapping reveals extensive structural variation in the human genome,” Science, 2007, 318(5849):420-426.
Kozlov et al., “A High-Complexity Multiplexed Solution-Phase Assay for Profiling Protease Activity on Microarrays,” Comb. Chem. and High Throughput, 11: 24-35, 2008.
Kozlov et al., “A highly scalable peptide-based assay system for proteomics,” PLoS ONE, 2012, 7(6):e37441, 10 pages.
Kretschy et al., “Next-Generation o-Nitrobenzyl Photolabile Groups for Light-Directed Chemistry and Microarray Synthesis,” Angewandte Chemie International Edition, Jul. 2015, 54(29):8555-8559.
Kristensen et al., “High-Throughput Methods for Detection of Genetic Variation,” BioTechniques, Feb. 2001, 30(2):318-332.
Kurz et al., “cDNA—protein fusions: covalent protein—gene conjugates for the in vitro selection of peptides and proteins,” ChemBioChem., 2001, 2(9):666-72.
Kwok, “High-throughput genotyping assay approaches,” Pharmocogenomics, Feb. 2000, 1(1):95-100.
Lage et al., “Whole genome analysis of genetic alterations in small DNA samples using hyperbranched strand displacement amplification and array-CGH,” Genome Research, 2003, 13(2):294-307.
Lamture et al., “Direct detection of nucleic acid hybridization on the surface of a charge coupled device,” Nucleic Acid Res., Jun. 1994, 22(11):2121-5.
Landegren et al., “Reading bits of genetic information: methods for single-nucleotide polymorphism analysis,” Genome Res., Aug. 1998, 8(8):769-76.
Langdale et al., “A rapid method of gene detection using DNA bound to Sephacryl,” Gene, 1985, 36(3):201-210.
Larsen et al., “Characterization of a recombinantly expressed proteinase K-like enzyme from a psychrotrophic Serratia sp,” FEBS J., Jan. 2006, 273(1):47-60.
Lee et al., “Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues,” Nature Protocols, 2015, 10(3):442-458.
Leriche et al., “Cleavable linkers in chemical biology,” Bioorganic & Medicinal Chemistry, 2012, 20:571-582.
Lindstrom et al., “Miniaturization of biological assays—Overview on microwell devices for single-cell analyses,” Biochimica et Biophysica Acta, 2011, 1810:308-316.
Linnarsson, “Recent advances in DNA sequencing methods—general principles of sample preparation,” Experimental Cell Research, 2010, 316(8):1339-1343.
Liu et al., “High-Spatial-Resolution Multi-Omics Atlas Sequencing of Mouse Embryos via Deterministic Barcoding in Tissue,” BioRxiv, 2019, 55 pages.
Lizardi et al., “Mutation detection and single-molecule counting using isothermal rolling-circle amplification,” Nat. Genet., 1998, 19(3):225-232.
Lundberg et al., “Multiplexed homogeneous proximity ligation assays for high-throughput protein biomarker research in serological material,” Mol Cell Proteomics, 2011, 10(4):M110.004978, 11 pages.
MacBeath et al., “Printing proteins as microarrays for high-throughput function determination,” Science, Sep. 2000, 289(5485):1760-1763.
Marx, “Method of the Year: spatially resolved transcriptomics,” Nature Methods, 2021, 18(1):9-14.
Merritt et al., “Multiplex digital spatial profiling of proteins and RNA in fixed tissue,” Nat Biotechnol, May 2020, 38(5):586-599.
Metzker, “Sequencing technologies—the next generation,” Nature Reviews Genetics, 2010, 11(1):31-46.
Miller et al., “Basic concepts of microarrays and potential applications in clinical microbiology,” Clinical Microbiology Reviews, 2009, 22(4):611-633.
Mishra et al., “Three-dimensional genome architecture and emerging technologies: looping in disease,” Genome Medicine, 2017, 9(1):87, 14 pages.
Mitra et al., “Digital genotyping and haplotyping with polymerase colonies,” Proc. Natl. Acad. Sci. USA, May 2003, 100(10):5926-5931.
Mizusawa et al., “A bacteriophage lambda vector for cloning with BamHI and Sau3A,” Gene, 1982, 20(3):317-322.
Nawy, “Spatial transcriptomics”, Nature Methods, vol. 15, No. 1, 2018.
Nikiforov et al., “The use of 96-well polystyrene plates for DNA hybridization-based assays: an evaluation of different approaches to oligonucleotide immobilization,” Anal Biochem, May 1995, 227(1):201-9.
Nowak et al., “Entering the Postgenome Era,” Science, 1995, 270(5235):368-71.
Pemov et al., “DNA analysis with multiplex microarray-enhanced PCR,” Nucl. Acids Res., Jan. 2005, 33(2):e11, 9 pages.
Perler et al., “Intervening sequences in an Archaea DNA polymerase gen,” Proc Natl Acad Sci USA, Jun. 1992, 89(12):5577-5581.
Petterson et al., “Generations of sequencing technologies,” Genomics, 2009, 93(2):105-111.
Polsky-Cynkin et al., “Use of DNA immobilized on plastic and agarose supports to detect DNA by sandwich hybridization,” Clin. Chem., 1985, 31(9):1438-1443.
Ranki et al., “Sandwich hybridization as a convenient method for the detection of nucleic acids in crude samples,” Gene, 1983, 21(1-2):77-85.
Reinartz et al., “Massively parallel signature sequencing (MPSS) as a tool for in-depth quantitative gene expression profiling in all organisms,” Brief Funct Genomic Proteomic, Feb. 2002, 1(1):95-104.
Ristova et al., “Study of hydrogenated amorphous silicon thin films as a potential sensor for He—Ne laser light detection,” Applied Surface Science, Sep. 2003, 218(1-4):44-53.
Rodriques et al., “Slide-seq: A scalable technology for measuring genome-wide expression at high spatial resolution,” Science, 2019, 363(6434):1463-1467.
Ronaghi et al., “A sequencing method based on real-time pyrophosphate,” Science, Jul. 1998, 281(5375):363-365.
Ronaghi et al., “Real-time DNA sequencing using detection of pyrophosphate release,” Analytical Biochemistry, Nov. 1996, 242(1):84-89.
Ronaghi, “Pyrosequencing sheds light on DNA sequencing,” Genome Res, Jan. 2001, 11(1):3-11.
Salmén et al., “Barcoded solid-phase RNA capture for Spatial Transcriptomics profiling in mammalian tissue sections,” Nature Protocols, Oct. 2018, 13(11):2501-2534.
Saxonov et al., “10x Genomics, Mastering Biology to Advance Human Health,” PowerPoint, 10x, 2020, 41 pages.
Schena et al., “Quantitative monitoring of gene expression patterns with a complementary DNA microarray,” Science, Oct. 1995, 270(5235):467-470.
Shalon et al., “A DNA microarray system for analyzing complex DNA samples using two-color fluorescent probe hybridization,” Genome Res., Jul. 1996, 6(7):639-45.
Stahl et al., “Visualization and analysis of gene expression in tissue sections by spatial transcriptomics,” Science, Jul. 2016, 353(6294):78-82.
Stahl et al., “Visualization and analysis of gene expression in tissue sections by spatial transcriptomics,” Supplementary Materials, Science, Jul. 2016, 353(6294):78-82, 41 pages.
Stimpson et al., “Real-time detection of DNA hybridization and melting on oligonucleotide arrays by using optical wave guides,” Proc Natl Acad Sci USA, Jul. 1995, 92(14):6379-83.
Strell et al., “Placing RNA in context and space—methods for spatially resolved transcriptomics,” The FEBS Journal, 2019, 286(8):1468-1481.
Tijssen et al., “Overview of principles of hybridization and the strategy of nucleic acid assays” in Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Acid Probes, 1993, 24(Chapter 2), 65 pages.
Trejo et al., “Extraction-free whole transcriptome gene expression analysis of FFPE sections and histology-directed subareas of tissue,” PLoS ONE, Feb. 2019, 14(2):e0212031, 22 pages.
Twyman et al., “Techniques Patents for SNP Genotyping,” Pharmacogenomics, Jan. 2003, 4(1):67-79.
Valley et al., “Optoelectronic tweezers as a tool for parallel single-cell manipulation and stimulation,” IEEE Trans Biomed Circuits Syst., Dec. 2009, 3(6):424-31.
Van Gelder et al., “Amplified RNA synthesized from limited quantities of heterogeneous cDNA,” Proc. Natl. Acad. Sci. USA, 1990, 87(5):1663-1667.
Vasiliskov et al., “Fabrication of microarray of gel-immobilized compounds on a chip by copolymerization,” Biotechniques, Sep. 1999, 27(3):592-606.
Vickovic et al., “High-definition spatial transcriptomics for in situ tissue profiling,” Nat Methods, Oct. 2019, 16(10):987-990.
Vogelstein et al., “Digital PCR,” Proceedings of the National Academy of Sciences, Aug. 1999, 96(16):9236-9241.
Walker et al., “Strand displacement amplification—an isothermal, in vitro DNA amplification technique,” Nucleic Acids Research, 1992, 20(7):1691-1696.
Wang et al., “Imaging-based pooled CRISPR screening reveals regulators of IncRNA localization,” Proc Natl Acad Sci USA, May 2019, 116(22):10842-10851.
Wang et al., “High-fidelity mRNA amplification for gene profiling,” Nature Biotechnology, Apr. 2000, 18(4):457-459.
Worthington et al., “Cloning of random oligonucleotides to create single-insert plasmid libraries,” Anal Biochem, 2001, 294(2):169-175.
Yershov et al., “DNA analysis and diagnostics on oligonucleotide microchips,” Proc. Natl. Acad. Sci. USA, May 1996, 93(10):4913-4918.
Zhu et al., “Reverse transcriptase template switching: a SMART approach for full-length cDNA library construction,” Biotechniques, Apr. 2001, 30(4):892-897.
Provisional Applications (1)
Number Date Country
62985103 Mar 2020 US