Methods of making gene expression libraries

Information

  • Patent Grant
  • 11891654
  • Patent Number
    11,891,654
  • Date Filed
    Wednesday, February 24, 2021
    3 years ago
  • Date Issued
    Tuesday, February 6, 2024
    3 months ago
Abstract
Provided herein are methods of determining a location of a target nucleic acid in a biological sample.
Description
BACKGROUND

Cells within a tissue of a subject 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, and signaling and cross-talk with other cells in the tissue.


Spatial heterogeneity has been previously studied using techniques that only provide data for a small handful of analytes in the context of an intact tissue or a portion of a tissue, or provide a lot of analyte data for single cells, but fail to provide information regarding the position of the single cell in a parent biological sample (e.g., tissue sample).


SUMMARY

This application is based on the discovery of a method of making a spatial 5′ gene expression library for spatial analysis of target analytes, including long target analytes e.g., VDJ rearranged T-cell receptors or immunoglobulins.


Provided herein are methods of identifying a location of a target nucleic acid in a permeabilized biological sample, the method comprising: (a) generating a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence, wherein the step of generating the cDNA molecule occurs within the permeabilized biological sample; (b) ligating a second adaptor sequence to a 3′ end of the cDNA molecule, wherein the step of ligating is performed within the biological sample; (c) after step (b), releasing the cDNA molecule from the target nucleic acid, such that the cDNA contacts an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence ligated to the cDNA; (d) after step (c), extending a 3′ end of the capture probe using the cDNA as a template; and (e) determining (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) all or a part of the sequence of the spatial barcode, or a complement thereof, and using the determined sequences of (i) and (ii) to identify the location of the target nucleic acid in the permeabilized biological sample.


Also provided herein are methods of identifying a location of a target nucleic acid in a permeabilized biological sample, the method comprising: (a) generating a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence, wherein the step of generating the cDNA molecule occurs within the permeabilized biological sample; (b) extending a 3′ end of the cDNA molecule to include a second adaptor sequence, wherein the step of extending is performed within the biological sample; (c) releasing the cDNA molecule from the target nucleic acid, such that the cDNA contacts an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence; (d) extending a 3′ end of the capture probe using the cDNA as a template; and (e) determining (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) the sequence of the spatial barcode, or a complement thereof, and using the determined sequences of (i) and (ii) to identify the location of the target nucleic acid in the permeabilized biological sample. In some embodiments, step (b) can occur simultaneously with step (a).


In some embodiments of any of the methods described herein, steps (a) through (c) are performed when the biological sample is disposed on the array.


In some embodiments of any of the methods described herein, step (a) is performed when the biological sample is not disposed on the array and step (b) is performed when the biological sample is disposed on the array, and wherein the method further comprises between steps (a) and (b), a step of disposing the biological sample on the array.


In some embodiments of any of the methods described herein, steps (a) and (b) are performed when the biological sample is not disposed on the array, and wherein the method further comprises between steps (b) and (c), a step of disposing the biological sample on the array.


In some embodiments of any of the methods described herein, the sequence that is substantially complementary to a portion of the target nucleic acid present in the reverse transcription primer comprises a poly(T) sequence.


In some embodiments of any of the methods described herein, the sequence that is substantially complementary to a portion of the target nucleic acid present in the reverse transcription primer comprises a random sequence.


In some embodiments of any of the methods described herein, the second adaptor sequence is a template switching oligonucleotide (TSO).


In some embodiments of any of the methods described herein, the array comprises a slide.


In some embodiments of any of the methods described herein, a 5′ end of the capture probe is attached to the slide.


In some embodiments of any of the methods described herein, the array is a bead array.


In some embodiments of any of the methods described herein, a 5′ end of the capture probe is attached to a bead of the bead array.


In some embodiments of any of the methods described herein, the capture probe further comprises a unique molecular identifier (UMI).


In some embodiments of any of the methods described herein, the UMI is positioned 5′ relative to the capture domain in the capture probe.


In some embodiments of any of the methods described herein, the determining in step (e) comprises sequencing (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) all or a part of the sequence of the spatial barcode, or a complement thereof.


In some embodiments of any of the methods described herein, the sequencing is high throughput sequencing.


In some embodiments of any of the methods described herein, the sequencing is sequencing by hybridization.


In some embodiments of any of the methods described herein, the target nucleic acid is RNA.


In some embodiments of any of the methods described herein, the RNA is an mRNA.


In some embodiments of any of the methods described herein, the permeabilized biological sample is a permeabilized tissue section.


In some embodiments of any of the methods described herein, the permeabilized tissue section is a permeabilized formalin-fixed and paraffin-embedded (FFPE) tissue section.


Some embodiments of any of the methods described herein further comprises a step of imaging the biological sample.


In some embodiments of any of the methods described herein, the step of imaging is performed prior to step (a).


In some embodiments of any of the methods described herein, the step of imaging is performed between steps (b) and (c).


Some embodiments of any of the methods described herein further comprises, between steps (b) and (c), a step of freezing and thawing the permeabilized biological sample.


Some embodiments of any of the methods described herein further comprises, between steps (b) and (c), a step of sectioning the permeabilized biological sample.


In some embodiments of any of the methods described herein, the step of sectioning the permeabilized biological sample is performed using cryosectioning.


Some embodiments of any of the methods described herein further comprises, prior to step (a), a step of permeabilizing the biological sample.


In some embodiments of any of the methods described herein, the performance of step (a) comprises introducing a reverse transcriptase, dNTPs, and the reverse transcription primer into the permeabilized biological sample.


In some aspects, provided herein are kits comprising: a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence; a reverse transcriptase; and an oligonucleotide comprising a second adaptor sequence or a complement thereof.


In some embodiments of any of the kits provided herein, the kit further comprises a ligase.


In some embodiments of any of the kits provided herein, the reverse transcriptase is a reverse transcriptase with terminal transferase activity.


In some embodiments of any of the kits provided herein, the second adaptor sequence or the complement thereof is a TSO or a complement thereof.


In some embodiments of any of the kits provided herein, the kit further comprises an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence.


In another aspect, provided herein are nucleic acids comprising, in the 5′ to 3′ direction: a spatial barcode; a sequence complementary to a second adaptor sequence; a sequence present in a target nucleic acid; and a sequence complementary to a first adaptor sequence.


In another aspect, provided herein are nucleic acids comprising, in the 3′ to 5′ direction: a complement of a spatial barcode; a second adaptor sequence; a sequence complementary to a sequence present in a target nucleic acid; and a first adaptor sequence.


In some embodiments of any of the nucleic acids provided herein, the second adaptor sequence comprises a TSO.


In some embodiments of any of the nucleic acids provided herein, the second adaptor sequence comprises a complement of a TSO.


In some embodiments of any of the nucleic acids provided herein, the first adaptor sequence is a reverse transcriptase primer.


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. 1A shows a workflow schematic illustrating exemplary, non-limiting steps for in-situ cDNA synthesis and capturing.



FIG. 1B shows a workflow schematic illustrating exemplary, non-limiting steps for building a 5′ spatial gene expression library.



FIG. 2A shows a workflow schematic illustrating exemplary, non-limiting steps for in-situ cDNA synthesis and sample handling.



FIG. 2B shows a workflow schematic illustrating exemplary, non-limiting steps for building a 5′ spatial gene expression library.



FIG. 3A shows a workflow schematic illustrating exemplary, non-limiting steps for synthesizing a cDNA molecule.



FIG. 3B shows a workflow schematic illustrating exemplary, non-limiting steps for cDNA binding to an attached capture probe and the extension of the capture probe using the cDNA molecule as a template, and the generation of a second strand complementary to the extended capture probe.



FIG. 3C shows a workflow schematic illustrating exemplary, non-limiting, non-exhaustive steps for building a 5′ spatial gene expression library.





DETAILED DESCRIPTION

In some cases, spatial analysis methods can be carried out by permeabilizing a biological sample, capturing analytes (e.g., nucleic acids (e.g., mRNA)) or intermediate agents on an array, and performing reverse transcription and sequencing steps to identify the location of one or more analytes from the biological sample. Many capture protocols rely on the use of a poly(A) tail, either natural or introduced. Several challenges can arise from these protocols. For example, there may or may not be biased in the analytes or intermediate agents that are able to migrate from a biological sample to the array. As another example, capture by the poly(A) tail can lead to a 3′ bias in gene expression libraries generated from mRNAs due to limitations of some steps of the process. Provided herein are methods that can, in some cases, address one or both of these challenges.


Provided herein are methods of identifying a location of a target nucleic acid in a permeabilized biological sample, the method comprising: (a) generating a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence, wherein the step of generating the cDNA molecule occurs within the permeabilized biological sample; (b) ligating a second adaptor sequence to a 3′ end of the cDNA molecule, wherein the step of ligating is performed within the biological sample; (c) after step (b), releasing the cDNA molecule from the target nucleic acid, such that the cDNA contacts an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence ligated to the cDNA; (d) after step (c), extending a 3′ end of the capture probe using the cDNA as a template; and (e) determining (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) all or a part of the sequence of the spatial barcode, or a complement thereof, and using the determined sequences of (i) and (ii) to identify the location of the target nucleic acid in the permeabilized biological sample. Non-limiting aspects of these methods are described herein.


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 domain can include a sequence that is significantly complementary to an analyte, a complement thereof, or a portion thereof (e.g., a capture domain can include a poly-T sequence). In some embodiments, a capture domain can include a sequence that is significantly complementary to a sequence introduced to the analyte before capture (e.g., a capture domain can include a sequence complementary to a functional domain and/or an adaptor sequence (e.g., a template switching oligonucleotide sequence)). 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, genetic material is amplified by reverse transcription polymerase chain reaction (RT-PCR). The desired reverse transcriptase activity can be provided by one or more distinct reverse transcriptase enzymes (i.e., RNA dependent DNA polymerases), suitable examples of which include, but are not limited to: M-MLV, MuLV, AMV, HIV, ArrayScript™, MultiScribe™, ThermoScript™, and SuperScript® I, II, III, and IV enzymes. “Reverse transcriptase” includes not only naturally occurring enzymes, but all such modified derivatives thereof, including also derivatives of naturally-occurring reverse transcriptase enzymes.


In addition, reverse transcription can be performed using sequence-modified derivatives or mutants of M-MLV, MuLV, AMV, and HIV reverse transcriptase enzymes, including mutants that retain at least some of the functional, e.g., reverse transcriptase, activity of the wild-type sequence. The reverse transcriptase enzyme can be provided as part of a composition that includes other components, e.g., stabilizing components that enhance or improve the activity of the reverse transcriptase enzyme, such as RNase inhibitor(s), inhibitors of DNA-dependent DNA synthesis, e.g., actinomycin D. Many sequence-modified derivative or mutants of reverse transcriptase enzymes, e.g., M-MLV, and compositions including unmodified and modified enzymes are commercially available, e.g., ArrayScript™, MultiScribe™, ThermoScript™, and SuperScript® I, II, III, and IV enzymes.


Certain reverse transcriptase enzymes (e.g., Avian Myeloblastosis Virus (AMV) Reverse Transcriptase and Moloney Murine Leukemia Virus (M-MuLV, MMLV) Reverse Transcriptase) can synthesize a complementary DNA strand using both RNA (cDNA synthesis) and single-stranded DNA (ssDNA) as a template. Thus, in some embodiments, the reverse transcription reaction can use an enzyme (reverse transcriptase) that is capable of using both RNA and ssDNA as the template for an extension reaction, e.g., an AMV or MMLV reverse transcriptase.


In some embodiments, the quantification of RNA and/or DNA is carried out by real-time PCR (also known as quantitative PCR or qPCR), using techniques well known in the art, such as but not limited to “TAQMAN™”, or dyes such as “SYBR®”, or on capillaries (“LightCycler® Capillaries”). In some embodiments, the quantification of genetic material is determined by optical absorbance and with real-time PCR. In some embodiments, the quantification of genetic material is determined by digital PCR. In some embodiments, the genes analyzed can be compared to a reference nucleic acid extract (DNA and RNA) corresponding to the expression (mRNA) and quantity (DNA) in order to compare expression levels of the target nucleic acids.


A “template switching oligonucleotide” (TSO) is an oligonucleotide that hybridizes to untemplated nucleotides added by a reverse transcriptase (e.g., enzyme with terminal transferase activity) during reverse transcription. In some embodiments, a template switching oligonucleotide hybridizes to untemplated poly(C) nucleotides added by a reverse transcriptase. In some embodiments, the template switching oligonucleotide adds a common 5′ sequence to full-length cDNA that is used for cDNA amplification.


In some embodiments, the template switching oligonucleotide adds a common sequence onto the 5′ end of the RNA being reverse transcribed. For example, a template switching oligonucleotide can hybridize to untemplated poly(C) nucleotides added onto the end of a cDNA molecule and provide a template for the reverse transcriptase to continue replication to the 5′ end of the template switching oligonucleotide, thereby generating full-length cDNA ready for further amplification. In some embodiments, once a full-length cDNA molecule is generated, the template switching oligonucleotide can serve as a primer in a cDNA amplification reaction.


In some embodiments, a template switching oligonucleotide is added before, contemporaneously with, or after a reverse transcription, or other terminal transferase-based reaction. In some embodiments, a template switching oligonucleotide or complement thereof is included in the capture probe. In some embodiments, the TSO, or complement thereof, in the capture probe serves as a capture domain. In certain embodiments, methods of sample analysis using template switching oligonucleotides can involve the generation of nucleic acid products from analytes of the tissue sample, followed by further processing of the nucleic acid products with the template switching oligonucleotide.


Template switching oligonucleotides can include a hybridization region and a template region. The hybridization region can include any sequence capable of hybridizing to the target sequence. In some embodiments, the hybridization region can, e.g., include a series of G bases to complement the overhanging C bases at the 3′ end of a cDNA molecule. The series of G bases can include 1 G base, 2 G bases, 3 G bases, 4 G bases, 5 G bases, or more than 5 G bases. The template sequence can include any sequence to be incorporated into the cDNA. In other embodiments, the hybridization region can include at least one base in addition to at least one G base. In other embodiments, the hybridization can include bases that are not a G base. In some embodiments, the template region includes at least 1 (e.g., at least 2, 3, 4, 5 or more) tag sequences and/or functional sequences. In some embodiments, the template region and hybridization region are separated by a spacer.


In some embodiments, the template regions include a barcode sequence. The barcode sequence can act as a spatial barcode and/or as a unique molecular identifier. In some embodiments, the template region can include a functional region, for example a region that can be used for amplification, a region that is complementary to a capture domain on a capture probe, etc. In some embodiments, the template region can include a barcode and/or a unique molecular identifier and/or a functional sequence and/or a capture domain sequence. Template switching oligonucleotides can include deoxyribonucleic acids; ribonucleic acids; modified nucleic acids including 2-aminopurine, 2,6-diaminopurine (2-amino-dA), inverted dT, 5-methyl dC, 2′-deoxyInosine, Super T (5-hydroxybutynl-2′-deoxyuridine), Super G (8-aza-7-deazaguanosine), locked nucleic acids (LNAs), unlocked nucleic acids (UNAs, e.g., UNA-A, UNA-U, UNA-C, UNA-G), Iso-dG, Iso-dC, 2′ fluoro bases (e.g., Fluoro C, Fluoro U, Fluoro A, and Fluoro G), or any combination of the foregoing.


In some embodiments, the length of a template switching oligonucleotide can be at least about 1, 2, 10, 20, 50, 75, 100, 150, 200, or 250 nucleotides or longer. In some embodiments, the length of a template switching oligonucleotide can be at most about 2, 10, 20, 50, 100, 150, 200, or 250 nucleotides or longer.


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 ligation 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. 2020/0277663.


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.


Spatial 5′ Gene Expression Libraries


Provided herein are methods of identifying a location of a target nucleic acid in a permeabilized biological sample, the method comprising: (a) generating a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence, wherein the step of generating the cDNA molecule occurs within the permeabilized biological sample; (b) ligating a second adaptor sequence to a 3′ end of the cDNA molecule, wherein the step of ligating is performed within the biological sample; (c) releasing the cDNA molecule from the target nucleic acid, such that the cDNA contacts an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence ligated to the cDNA; (d) extending a 3′ end of the capture probe using the cDNA as a template; and (e) determining (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) all or a part of the sequence of the spatial barcode, or a complement thereof, and using the determined sequences of (i) and (ii) to identify the location of the target nucleic acid in the permeabilized biological sample.


Also provided herein are methods of identifying a location of a target nucleic acid in a permeabilized biological sample, the method comprising: (a) generating a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence, wherein the step of generating the cDNA molecule occurs within the permeabilized biological sample; (b) extending a 3′ end of the cDNA molecule to include a second adaptor sequence, wherein the step of extending is performed within the biological sample; (c) releasing the cDNA molecule from the target nucleic acid, such that the cDNA contacts an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence ligated to the cDNA; (d) extending a 3′ end of the capture probe using the cDNA as a template; and (e) determining (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) all or a part of the sequence of the spatial barcode, or a complement thereof, and using the determined sequences of (i) and (ii) to identify the location of the target nucleic acid in the permeabilized biological sample. In some such embodiments, the method can further comprise hybridizing a template switching oligonucleotide (TSO) to the cDNA molecule. Therefore, in some embodiments, (b) comprises extending a 3′ end of the cDNA molecule to include a complement of a TSO. In some cases, the TSO can be added to the sample at the same time as the reverse transcriptase primer.


In some embodiments, steps (a) through (c) are performed when the biological sample is disposed on the array. In some embodiments, step (a) is performed when the biological sample is not disposed on the array and step (b) is performed when the biological sample is disposed on the array, and wherein the method further comprises between steps (a) and (b), a step of disposing the biological sample on the array. In some embodiments, steps (a) and (b) are performed when the biological sample is not disposed on the array, and wherein the method further comprises between steps (b) and (c), a step of disposing the biological sample on the array.


In some embodiments of any of the methods described herein, the biological sample can be any of the exemplary permeabilized biological samples described herein (e.g., a permeabilized tissue sample, e.g., a permeabilized permeabilized tissue section), or any of the same described in, e.g., Section (I)(d) (e.g., (I)(d)(i) and/or (I)(d)(ii)(13)) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Some embodiments described herein can optionally further include a step of permeabilizing the biological sample (e.g., using any of the exemplary methods and agents for permeabilizing a biological sample described herein). In some embodiments, the target nucleic acid is RNA (e.g., mRNA).


In some embodiments, the reverse transcription primer can have a total of about 10 nucleotides to about 250 nucleotides (e.g., about 10 nucleotides to about 225 nucleotides, about 10 nucleotides to about 200 nucleotides, about 10 nucleotides to about 175 nucleotides, about 10 nucleotides to about 150 nucleotides, about 10 nucleotides to about 125 nucleotides, about 10 nucleotides to about 100 nucleotides, about 10 nucleotides to about 80 nucleotides, about 10 nucleotides to about 60 nucleotides, about 10 nucleotides to about 40 nucleotides, about 10 nucleotides to about 20 nucleotides, about 20 nucleotides to about 250 nucleotides, about 20 nucleotides to about 225 nucleotides, about 20 nucleotides to about 200 nucleotides, about 20 nucleotides to about 175 nucleotides, about 20 nucleotides to about 150 nucleotides, about 20 nucleotides to about 125 nucleotides, about 20 nucleotides to about 100 nucleotides, about 20 nucleotides to about 80 nucleotides, about 20 nucleotides to about 60 nucleotides, about 20 nucleotides to about 40 nucleotides, about 40 nucleotides to about 250 nucleotides, about 40 nucleotides to about 225 nucleotides, about 40 nucleotides to about 200 nucleotides, about 40 nucleotides to about 175 nucleotides, about 40 nucleotides to about 150 nucleotides, about 40 nucleotides to about 125 nucleotides, about 40 nucleotides to about 100 nucleotides, about 40 nucleotides to about 80 nucleotides, about 40 nucleotides to about 60 nucleotides, about 60 nucleotides to about 250 nucleotides, about 60 nucleotides to about 225 nucleotides, about 60 nucleotides to about 200 nucleotides, about 60 nucleotides to about 175 nucleotides, about 60 nucleotides to about 150 nucleotides, about 60 nucleotides to about 125 nucleotides, about 60 nucleotides to about 100 nucleotides, about 60 nucleotides to about 80 nucleotides, about 80 nucleotides to about 250 nucleotides, about 80 nucleotides to about 225 nucleotides, about 80 nucleotides to about 200 nucleotides, about 80 nucleotides to about 175 nucleotides, about 80 nucleotides to about 150 nucleotides, about 80 nucleotides to about 125 nucleotides, about 80 nucleotides to about 100 nucleotides, about 100 nucleotides to about 250 nucleotides, about 100 nucleotides to about 225 nucleotides, about 100 nucleotides to about 200 nucleotides, about 100 nucleotides to about 175 nucleotides, about 100 nucleotides to about 150 nucleotides, about 100 nucleotides to about 125 nucleotides, about 125 nucleotides to about 250 nucleotides, about 125 nucleotides to about 225 nucleotides, about 125 nucleotides to about 200 nucleotides, about 125 nucleotides to about 175 nucleotides, about 125 nucleotides to about 150 nucleotides, about 150 nucleotides to about 250 nucleotides, about 150 nucleotides to about 225 nucleotides, about 150 nucleotides to about 200 nucleotides, about 150 nucleotides to about 175 nucleotides, about 175 nucleotides to about 250 nucleotides, about 175 nucleotides to about 225 nucleotides, about 175 nucleotides to about 200 nucleotides, about 200 nucleotides to about 250 nucleotides, about 200 nucleotides to about 225 nucleotides, or about 225 nucleotides to about 250 nucleotides).


In some embodiments of any of the methods described herein, the first adaptor sequence has a total of about 5 nucleotides to about 125 nucleotides (e.g., about 5 nucleotides to about 100 nucleotides, about 5 nucleotides to about 90 nucleotides, about 5 nucleotides to about 80 nucleotides, about 5 nucleotides to about 70 nucleotides, about 5 nucleotides to about 60 nucleotides, about 5 nucleotides to about 50 nucleotides, about 5 nucleotides to about 45 nucleotides, about 5 nucleotides to about 40 nucleotides, about 5 nucleotides to about 35 nucleotides, about 5 nucleotides to about 30 nucleotides, about 5 nucleotides to about 25 nucleotides, about 5 nucleotides to about 20 nucleotides, about 5 nucleotides to about 15 nucleotides, about 5 nucleotides to about 10 nucleotides, about 10 nucleotides to about 125 nucleotides, about 10 nucleotides to about 100 nucleotides, about 10 nucleotides to about 90 nucleotides, about 10 nucleotides to about 80 nucleotides, about 10 nucleotides to about 70 nucleotides, about 10 nucleotides to about 60 nucleotides, about 10 nucleotides to about 50 nucleotides, about 10 nucleotides to about 45 nucleotides, about 10 nucleotides to about 40 nucleotides, about 10 nucleotides to about 35 nucleotides, about 10 nucleotides to about 30 nucleotides, about 10 nucleotides to about 25 nucleotides, about 10 nucleotides to about 20 nucleotides, about 10 nucleotides to about 15 nucleotides, about 20 nucleotides to about 125 nucleotides, about 20 nucleotides to about 100 nucleotides, about 20 nucleotides to about 90 nucleotides, about 20 nucleotides to about 80 nucleotides, about 20 nucleotides to about 70 nucleotides, about 20 nucleotides to about 60 nucleotides, about 20 nucleotides to about 50 nucleotides, about 20 nucleotides to about 45 nucleotides, about 20 nucleotides to about 40 nucleotides, about 20 nucleotides to about 35 nucleotides, about 20 nucleotides to about 30 nucleotides, about 20 nucleotides to about 25 nucleotides, about 30 nucleotides to about 125 nucleotides, about 30 nucleotides to about 100 nucleotides, about 30 nucleotides to about 90 nucleotides, about 30 nucleotides to about 80 nucleotides, about 30 nucleotides to about 70 nucleotides, about 30 nucleotides to about 60 nucleotides, about 30 nucleotides to about 50 nucleotides, about 30 nucleotides to about 45 nucleotides, about 30 nucleotides to about 40 nucleotides, about 30 nucleotides to about 35 nucleotides, about 40 nucleotides to about 125 nucleotides, about 40 nucleotides to about 100 nucleotides, about 40 nucleotides to about 90 nucleotides, about 40 nucleotides to about 80 nucleotides, about 40 nucleotides to about 70 nucleotides, about 40 nucleotides to about 60 nucleotides, about 40 nucleotides to about 50 nucleotides, about 40 nucleotides to about 45 nucleotides, about 50 nucleotides to about 125 nucleotides, about 50 nucleotides to about 100 nucleotides, about 50 nucleotides to about 90 nucleotides, about 50 nucleotides to about 80 nucleotides, about 50 nucleotides to about 70 nucleotides, about 50 nucleotides to about 60 nucleotides, about 60 nucleotides to about 125 nucleotides, about 60 nucleotides to about 100 nucleotides, about 60 nucleotides to about 90 nucleotides, about 60 nucleotides to about 80 nucleotides, about 60 nucleotides to about 70 nucleotides, about 70 nucleotides to about 125 nucleotides, about 70 nucleotides to about 100 nucleotides, about 70 nucleotides to about 90 nucleotides, about 70 nucleotides to about 80 nucleotides, about 80 nucleotides to about 125 nucleotides, about 80 nucleotides to about 100 nucleotides, about 80 nucleotides to about 90 nucleotides, about 90 nucleotides to about 125 nucleotides, about 90 nucleotides to about 100 nucleotides, or about 100 nucleotides to about 125 nucleotides). In some embodiments, the first adaptor sequence can be any predetermined sequence. In some embodiments, the first adaptor sequence does not encode a polypeptide and/or can be non-naturally occurring sequence.


In some embodiments the sequence that is substantially complementary (e.g., at least 80%, at least 85%, at least 90%, at least 95%, at least 99%, or 100% complementary) to a portion of the sequence of the target nucleic acid (that is present in the reverse transcription primer) can have a total of about 10 nucleotides to about 125 nucleotides (or any of the subranges of this range described herein). In some embodiments, the sequence that is substantially complementary to a portion of the sequence of the target nucleic acid can be a random sequence. In some embodiments, the sequence that is substantially complementary to a portion of the sequence of the target nucleic acid can include a poly(T) oligonucleotide sequence (e.g., at least 5 contiguous Ts, at least 10 continguous Ts, or at least 15 contiguous Ts).


In some embodiments, the step of generating a cDNA molecule including a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer can include contacting a permeabilized biological sample with a reverse transcriptase (e.g., any of the exemplary reverse transcriptases described herein or known in the art), dNTPs, and the reverse transcription primer (e.g., any of the exemplary reverse transcription primers described herein). A variety of kits including a reverse transcriptase and dNTPs are commercially available. Non-limiting examples of conditions for generating a cDNA molecule are described herein, and additional examples of conditions for generating a cDNA molecule are known in the art.


In some embodiments of any of the methods described herein, the second adaptor sequence (e.g., ligated to a 3′ end of the generated cDNA molecule (performed within the biological sample), or included in the generated cDNA molecule via extension of a 3′ end of the cDNA molecule) can have a total of about 5 nucleotides to about 125 nucleotides (or any of the subranges of this range described herein). In some embodiments, the second adaptor sequence can be any predetermined sequence. In some embodiments, the second adaptor sequence does not encode a polypeptide and/or can be non-naturally occurring sequence. In some embodiments, the first and second adaptor sequences include different sequences. In some embodiments, the second adaptor sequence can be a template switching oligonucleotide (TSO) (e.g., any of the exemplary TSOs described herein), or a complement thereof. In some embodiments, the second adaptor sequence includes a sequence that is substantially complementary (e.g., at least 80%, at least 85%, at least 90%, at least 92%, at least 94%, at least 96%, at least 98%, at least 99%, or 100% complementary) to a sequence in the capture domain of the capture probe.


In some embodiments, the step of ligating the second adaptor sequence to a 3′ end of the generated cDNA molecule (performed within the biological sample) can be performed using any of the ligation methods described herein or known in the art. A wide variety of different methods can be used for ligating nucleic acid molecules, including (but not limited to) “sticky-end” and “blunt-end” ligations. Additionally, single-stranded ligation can be used to perform proximity ligation on a single-stranded nucleic acid molecule. Sticky-end proximity ligations involve the hybridization of complementary single-stranded sequences between the two nucleic acid molecules to be joined, prior to the ligation event itself. Blunt-end ligations generally do not include hybridization of complementary regions from each nucleic acid molecule because both nucleic acid molecules lack a single-stranded overhang at the site of ligation. In some embodiments, DNA ligase activity can be provided by one or more distinct DNA ligase enzymes. In some embodiments, the DNA ligase enzyme is from a bacterium, e.g., the DNA ligase enzyme is a bacterial DNA ligase enzyme. In some embodiments, the DNA ligase enzyme is from a virus (e.g., a bacteriophage). For instance, the DNA ligase can be T4 DNA ligase. Other enzymes appropriate for the ligation step include, but are not limited to, Tth DNA ligase, Taq DNA ligase, Thermococcus sp. (strain 9oN) DNA ligase (9oN™ DNA ligase, available from New England Biolabs, Ipswich, Mass.), and Ampligase® (available from Lucigen, Middleton, Wis.). Derivatives, e.g., sequence-modified derivatives, and/or mutants thereof, can also be used.


For example, the step of ligating can be performed by contacting the permeabilized biological sample with a ligase and the second adaptor sequence (e.g., a TSO), and optionally, any additional components required to accelerate the ligation reaction. In some embodiments, the methods can further include blocking the 5′ end of the generated cDNA molecule prior to the ligating step. Non-limiting examples of conditions for performing ligation are described herein, and additional examples of conditions for performing ligation are known in the art.


In some embodiments, extension of a 3′ end of a generated cDNA to include a second adaptor sequence can be performed using any appropriate methods, such as those described herein for a TSO. For example, the step of extension of a 3′ end of a generated cDNA molecule can include hybridizing an oligo comprising a complement of the second adaptor sequence to a portion of a 3′ end of the generated cDNA molecule, and extending the cDNA molecule to include the second adaptor sequence. In some cases, the terminal transferase activity of a reverse transcriptase enzyme will result in a polymononucleotide sequence (e.g., a poly(C) sequence) at the 3′ end of a generated cDNA molecule, and the oligo comprising a complement of the second adaptor sequence can further include a complementary polymononucleotide sequence (e.g., a poly(G) sequence) that allows for hybridization to the polymononucleotide sequence of the cDNA molecule. The hybridized oligo comprising the complement of the second adaptor sequence can then be used as a template for extending the 3′ end of the generated cDNA molecule to include the second adaptor sequence. In some cases, the oligo comprising a complement of the second adaptor sequence is added to the sample at the same time as the reverse transcription primer.


In some embodiments, the releasing of the cDNA molecule from the target nucleic acid can be performed by using heat or a chemical denaturant (e.g., KOH).


In some embodiments of any of the methods described herein, the array can be any of the types of arrays described herein. For example, the array includes a slide. In some embodiments, the capture probe is attached to the slide (e.g., by its 5′ end).


In some embodiments, the array is a bead array. In some embodiments, a 5′ end of the capture probe is attached to a bead of the bead array.


In some embodiments, the capture probe further comprises a unique molecular identifier (UMI) (e.g., a UMI positioned 5′ relative to the capture domain the capture probe).


In some embodiments, the determining in step (e) comprises sequencing (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) all or a part of the sequence of the spatial barcode, or a complement thereof. In some embodiments, the sequencing is high throughput sequencing, sequencing by hybridization, or any of the other methods for sequencing described herein or known in the art. For example, sequencing can involve one or more of nucleic acid amplification, the ligation or addition of one or more sequencing adaptors, cleavage of the capture probe from the array, extension of the capture probe using the bound cDNA as a template, and generating a single-stranded nucleic acid comprising a sequence that is complementary to the extended capture probe. Non-limiting methods for determining the sequence of (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, or (ii) all or a part of the sequence of the spatial barcode, or a complement thereof, are described herein or are known in the art.


In some embodiments, the methods can optionally further include a step of imaging the biological sample (e.g., using any of the exemplary imaging methods described herein or known in the art). In some embodiments, the imaging is performed prior to step (a). In some embodiments, the imaging is performed between steps (b) and (c).


In some embodiments, the method further includes, between steps (b) and (c), a step of freezing and thawing the permeabilized biological sample. In some embodiments, the method can further include, between steps (b) and (c), a step of sectioning (e.g., cryosectioning) the permeabilized biological sample.


EXEMPLARY EMBODIMENTS


FIG. 1A is an exemplary diagram showing, from left to right, the hybridization of a reverse transcription primer to a target nucleic acid, e.g., an mRNA, within a permeabilized biological sample; the generation of a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid, and the addition/ligation of a second adaptor sequence (e.g., a template switching oligonucleotide, or a complement thereof) to the 3′ end of the cDNA molecule within the permeabilized biological sample; and releasing of the cDNA molecule from the target nucleic acid and contacting the released cDNA molecule to an array (e.g., any of the exemplary arrays described herein) comprising a capture probe for performance of additional steps (e.g., any of the exemplary additional steps described herein). The second adaptor sequence added (e.g., via extension)/ligated to the cDNA specifically binds to the capture probe.



FIG. 1B is an exemplary workflow showing the generation of a gene expression library and the identification of the location of a target nucleic acid in a biological sample, for example following a target analyte capture workflow as shown pictorially in FIG. 1A. Specifically, a biological sample, e.g., a tissue sample, is fixed, stained, and imaged. Any of the exemplary methods described herein or known in the art can be used to fix, stain, and/or image the biological sample. In some embodiments, the biological sample can be a formalin-fixed and paraffin-embedded (FFPE) tissue sample. In some embodiments, the biological sample is stained, e.g., using an H&E staining method. In some embodiments, the tissue sample is fixed, stained, and/or imaged for 5 minutes to about 5 hours, e.g., about 5 minutes to about 4.5 hours, about 5 minutes to about 4.0 hours, about 5 minutes to about 3.5 hours, about 5 minutes to about 3.0 hours, about 5 minutes to about 2.5 hours, about 5 minutes to about 2.0 hours, about 5 minutes to about 1.5 hours, about 5 minutes to about 1.0 hour, about 5 minutes to about 50 minutes, about 5 minutes to about 40 minutes, about 5 minutes to about 30 minutes, about 5 minutes to about 20 minutes, about 5 minutes to about 10 minutes, about 10 minutes to about 5 hours, about 10 minutes to about 4.5 hours, about 10 minutes to about 4.0 hours, about 10 minutes to about 3.5 hours, about 10 minutes to about 3.0 hours, about 10 minutes to about 2.5 hours, about 10 minutes to about 2.0 hours, about 10 minutes to about 1.5 hours, about 10 minutes to about 1.0 hour, about 10 minutes to about 50 minutes, about 10 minutes to about 40 minutes, about 10 minutes to about 30 minutes, about 10 minutes to about 20 minutes, about 20 minutes to about 5 hours, about 20 minutes to about 4.5 hours, about 20 minutes to about 4.0 hours, about 20 minutes to about 3.5 hours, about 20 minutes to about 3.0 hours, about 20 minutes to about 2.5 hours, about 20 minutes to about 2.0 hours, about 20 minutes to about 1.5 hours, about 20 minutes to about 1.0 hour, about 20 minutes to about 50 minutes, about 20 minutes to about 40 minutes, about 20 minutes to about 30 minutes, about 30 minutes to about 5 hours, about 30 minutes to about 4.5 hours, about 30 minutes to about 4.0 hours, about 30 minutes to about 3.5 hours, about 30 minutes to about 3.0 hours, about 30 minutes to about 2.5 hours, about 30 minutes to about 2.0 hours, about 30 minutes to about 1.5 hours, about 30 minutes to about 1.0 hour, about 30 minutes to about 50 minutes, about 30 minutes to about 40 minutes, about 1.0 hour to about 5 hours, about 1.0 hour to about 4.5 hours, about 1.0 hour to about 4.0 hours, about 1.0 hour to about 3.5 hours, about 1.0 hour to about 3.0 hours, about 1.0 hour to about 2.5 hours, about 1.0 hour to about 2.0 hours, about 1.0 hour to about 1.5 hours, about 1.5 hour to about 5 hours, about 1.5 hour to about 4.5 hours, about 1.5 hour to about 4.0 hours, about 1.5 hour to about 3.5 hours, about 1.5 hour to about 3.0 hours, about 1.5 hour to about 2.5 hours, about 1.5 hour to about 2.0 hours, about 2.0 hour to about 5 hours, about 2.0 hour to about 4.5 hours, about 2.0 hour to about 4.0 hours, about 2.0 hour to about 3.5 hours, about 2.0 hour to about 3.0 hours, about 2.0 hour to about 2.5 hours, about 2.5 hour to about 5 hours, about 2.5 hour to about 4.5 hours, about 2.5 hour to about 4.0 hours, about 2.5 hour to about 3.5 hours, about 2.5 hour to about 3.0 hours, about 3.0 hour to about 5 hours, about 3.0 hour to about 4.5 hours, about 3.0 hour to about 4.0 hours, about 3.0 hour to about 3.5 hours, about 3.5 hour to about 5 hours, about 3.5 hour to about 4.5 hours, about 3.5 hour to about 4.0 hours, about 4.0 hour to about 5 hours, about 4.0 hour to about 4.5 hours, or about 4.5 hour to about 5 hours.


After the fixation, staining and imaging of the biological sample, the biological sample is permeabilized. Permeabilization of the biological sample (e.g., a tissue sample) can be performed using any of the exemplary methods or exemplary reagents described herein, or in, e.g., Section (I)(d)(ii)(13) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. In some embodiments, the permeabilization of the biological sample (e.g., tissue sample) can be performed for about 1 minute to about 5 hours (e.g., about 1 minute to about 4.5 hours, about 1 minute to about 4.0 hours, about 1 minute to about 3.5 hours, about 1 minute to about 3.0 hours, about 1 minute to about 2.5 hours, about 1 minute to about 2.0 hours, about 1 minute to about 1.5 hours, about 1 minute to about 1.0 hour, about 1 minute to about 50 minutes, about 1 minute to about 40 minutes, about 1 minute to about 30 minutes, about 1 minute to about 20 minutes, about 1 minute to about 10 minutes, about 1 minute to about 5 minutes, about 10 minutes to about 5.0 hours, about 10 minutes to about 4.5 hours, about 10 minutes to about 4.0 hours, about 10 minutes to about 3.5 hours, about 10 minutes to about 3.0 hours, about 10 minutes to about 2.5 hours, about 10 minutes to about 2.0 hours, about 10 minutes to about 1.5 hours, about 10 minutes to about 1.0 hour, about 10 minutes to about 50 minutes, about 10 minutes to about 40 minutes, about 10 minutes to about 30 minutes, about 10 minutes to about 20 minutes, about 20 minutes to about 5.0 hours, about 20 minutes to about 4.5 hours, about 20 minutes to about 4.0 hours, about 20 minutes to about 3.5 hours, about 20 minutes to about 3.0 hours, about 20 minutes to about 2.5 hours, about 20 minutes to about 2.0 hours, about 20 minutes to about 1.5 hours, about 20 minutes to about 1.0 hour, about 20 minutes to about 50 minutes, about 20 minutes to about 40 minutes, about 20 minutes to about 30 minutes, about 30 minutes to about 5.0 hours, about 30 minutes to about 4.5 hours, about 30 minutes to about 4.0 hours, about 30 minutes to about 3.5 hours, about 30 minutes to about 3.0 hours, about 30 minutes to about 2.5 hours, about 30 minutes to about 2.0 hours, about 30 minutes to about 1.5 hours, about 30 minutes to about 1.0 hour, about 30 minutes to about 50 minutes, about 30 minutes to about 40 minutes, about 1.0 hour to about 5.0 hours, about 1.0 hour to about 4.5 hours, about 1.0 hour to about 4.0 hours, about 1.0 hour to about 3.5 hours, about 1.0 hour to about 3.0 hours, about 1.0 hour to about 2.5 hours, about 1.0 hour to about 2.0 hours, about 1.0 hour to about 1.5 hours, about 2.0 hours to about 5.0 hours, about 2.0 hours to about 4.5 hours, about 2.0 hours to about 4.0 hours, about 2.0 hours to about 3.5 hours, about 2.0 hours to about 3.0 hours, about 2.0 hours to about 2.5 hours, about 3.0 hours to about 5.0 hours, about 3.0 hours to about 4.5 hours, about 3.0 hours to about 4.0 hours, about 3.0 hours to about 3.5 hours, about 4.0 hours to about 5.0 hours, about 4.0 hours to about 4.5 hours, or about 4.5 hours to about 5.0 hours).


After permeabilization of the biological sample, the target nucleic acid in the permeabilized biological sample is contacted and hybridized with a reverse transcription primer (e.g., any of the reverse transcription primers described herein) to generate a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid.


In some embodiments, after the generation of the cDNA molecule, a second adaptor sequence is ligated to the 3′ end of the cDNA molecule. Any suitable adaptor sequence described herein can be ligated to the 3′ end of the cDNA molecule. In some embodiments, the second adaptor molecule is a template switching oligonucleotide (TSO).


In some embodiments, a 3′ end of the cDNA molecule is extended to include a second adaptor sequence. Any adaptor sequence described herein can be included to the 3′ end of the cDNA molecule. In some embodiments, the second adaptor molecule is a template switching oligonucleotide (TSO), or a complement thereof.


The generation of the cDNA molecule and the addition (e.g., via extension) or ligation of the second adaptor sequence can be performed over about 1 minute to about 2 hours (e.g., about 1 minute to about 1.5 hours, about 1 minute to about 1.0 hour, about 1 minute to about 40 minutes, about 1 minute to about 20 minutes, about 1 minute to about 10 minutes, about 10 minutes to about 2.0 hours, about 10 minutes to about 1.5 hours, about 10 minutes to about 1.0 hour, about 10 minutes to about 40 minutes, about 10 minutes to about 20 minutes, about 20 minutes to about 2.0 hours, about 20 minutes to about 1.5 hours, about 20 minutes to about 1.0 hour, about 20 minutes to about 40 minutes, about 40 minutes to about 2.0 hours, about 40 minutes to about 1.5 hours, about 40 minutes to about 1.0 hour, about 1.0 hour to about 2.0 hours, about 1 hour to about 1.5 hours, or about 1.5 hours to about 2.0 hours).


In some embodiments, the reverse transcription occurs within the permeabilized biological sample, e.g., a permeabilized tissue sample. In some embodiments, the permeabilization and the generation of the cDNA molecule occurs while the biological sample is disposed on the array.


After the generation of the cDNA molecule and the addition (e.g., via extension) or ligation of the second adaptor sequence, the cDNA molecule can be denatured/released from the target nucleic acid and the cDNA molecule is contacted with an array comprising a capture probe. The capture probe can include in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence ligated to the cDNA. When contacting with the array with the cDNA molecule, the attached capture probe captures the cDNA molecule using the capture domain, and the 3′ end of the capture probe is extended to add a sequence that is substantially complementary to the cDNA molecule sequence. In some embodiments, the methods can further include generating a single-stranded nucleic acid that includes a sequence that is substantially complementary to the extended capture probe. The optional steps of extending a 3′ end of the capture probe (using the specifically bound cDNA as a template) and generating a single-stranded nucleic acid including a sequence complementary to the extended capture probe can be performed for about 5 minutes to about 2 hours (or any of the subranges within this range described herein).


In some embodiments, a single-stranded nucleic acid including a sequence complementary to the extended capture probe can be denatured from the extended capture probe, and optionally, transferred to a different tube or container for performance of additional steps.


The single-stranded nucleic acid including a sequence complementary to the extended capture probe can be quantitated and/or sequenced or at least partially sequenced using any of the methods described herein, or described in, e.g., Sections (II)(a) or (II)(g) (e.g., (II)(g)(iv)) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663 or known in the art. In some embodiments, the quantitation and/or sequencing of the single-stranded nucleic acid including a sequence complementary to the extended capture probe can be quantitated and/or sequenced or at least partially sequenced for about 10 minutes to about 2 hours (or any of the subranges of this range described herein).


Following the denaturing of the single-stranded nucleic acid including a sequence complementary to the extended capture probe, the single-stranded nucleic acid including the sequence complementary to the extended capture probe can be subjected to amplification, fragmentation, end-repairing, A-tailing, adaptor ligation, sample index PCR, and the construction and quality control of a gene expression library, using any of the exemplary methods described herein, or described in, e.g., Sections (II)(a) or (II)(g) (e.g., (II)(g)(iv)) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.



FIG. 2A is an exemplary diagram showing, from left to right, the hybridization of a reverse transcription primer to a target nucleic acid, e.g., an mRNA, within a permeabilized biological sample, e.g., a whole tissue sample that has not been sectioned, cut or further fragmented; the generation of a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid, and the addition (e.g., via extension) or ligation of a second adaptor sequence (e.g., a template switching oligonucleotide, or a complement thereof) to the 3′ end of the cDNA molecule within the permeabilized biological sample, e.g., a whole tissue sample that has not been sectioned, cut or further fragmented; the fixation and/or flash-freezing of the biological sample, and the cryosectioning of the whole tissue sample for use in additional steps. When using a whole tissue sample that has not been sectioned, cut or further fragmented, the steps described in FIG. 2A are performed when the biological sample is not disposed on an array.



FIG. 2B is an exemplary workflow showing the generation of a gene expression library and the identification of the location of a target nucleic acid in a biological sample, for example following the target analyte capture workflow of FIG. 2A. Specifically, a biological sample, e.g., a whole tissue sample, is permeabilized. Any suitable permeabilization method described herein, or in, e.g., Section (I)(d)(ii)(13) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663, or known in the art can be used to permeabilize the whole tissue sample. In some embodiments, the biological sample is permeabilized for about 5 minutes to about 5 hours (or any of the subranges of this range described herein).


After permeabilization of the biological sample, the target nucleic acid in the permeabilized biological sample is contacted and annealed with a reverse transcription primer to generate a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid. After synthesis of the cDNA molecule, a second adaptor sequence is added (e.g., via extension) or ligated to the 3′ end of the cDNA molecule. Any suitable adaptor sequence described in the current application can be used to ligate to the 3′ cDNA molecule. In some embodiments, the second adaptor molecule is a template switching oligonucleotide (TSO), or a complement thereof. The generation of the cDNA molecule and the addition (e.g., via extension) or ligation of the second adaptor sequence can be performed for about 10 minutes to about 2 hours (or any of the subranges of this range described herein). In some embodiments, the synthesis of the cDNA molecule occurs within a permeabilized whole tissue sample.


Following the generation of the cDNA molecule, the biological sample, e.g., the whole tissue sample, can be fixed and/or flash-frozen. Any suitable methods described herein, or in, e.g., Section (I)(d)(1)-(I)(d)(4) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663, or known in the art can be used to fix and flash-freeze the tissue sample. In some embodiments, the biological sample, e.g., the whole tissue sample is formalin-fixed and paraffin-embedded (FFPE). In some embodiments, the biological sample, e.g., whole tissue sample, is flash-frozen using liquid nitrogen. The flash-frozen tissue sample is then sectioned for future steps. In some embodiments, the sectioning is performed using cryosectioning. In some embodiments, the methods further comprise a thawing step, after the cryosectioning.


After sectioning, the biological sample, e.g., tissue sample, can be stained, and imaged. Any of the methods described herein, or in, e.g., Section (I)(d)(6) or (II)(a)(i) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663, or known in the art can be used to stain and/or image the biological sample. In some embodiments, the biological sample is stained using an H&E staining method. In some embodiments, the tissue sample is stained and imaged for about 10 minutes to about 2 hours (or any of the subranges of this range described herein). Additional time may be needed for staining and imaging of different types of biological samples.


In some embodiments, the generation of the cDNA occurs within the permeabilized biological sample, e.g., a permeabilized tissue sample. In some embodiments, the permeabilization and the generation of the cDNA occurs while the biological sample is not disposed on the array.


After the generation of the cDNA molecule, the addition (e.g., via extension) or ligation of the second adaptor sequence to a 3′ end of the cDNA, and the tissue fixation, freezing, sectioning, staining, and imaging, the cDNA is released/denatured from the target nucleic acid and the cDNA is contacted with an array comprising a capture probe. The capture probe can include in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence ligated to the cDNA. When the array is contacted with the cDNA, the capture probe binds specifically to the cDNA via the capture domain, and the 3′ end of the capture probe is extended (using the specifically bound cDNA as a template) to add a sequence that is substantially complementary to the cDNA. The method can further include generating a single-stranded nucleic acid that is complementary to the extended capture probe. The denaturing of the cDNA from the target nucleic acid, the extension of the capture probe (using the specifically bound cDNA as a template), and optional generation of a single-stranded nucleic acid including a sequence that is complementary to the extended capture probe can be performed over 10 minutes to about 5 hours (or any of the subranges of this range described herein).


The single-stranded nucleic acid including a sequence that is complementary to the extended capture probe can be separated/denatured from the extended capture probe and optionally, transferred to a container (e.g., a strip tube) for the performance of additional steps.


The extended capture probe and/or a denatured/separated single-stranded nucleic acid including a sequence that is complementary to the extended capture probe can be quantitated and/or sequenced or at least partially sequenced using any of the methods described herein, or described in, e.g., Sections (II)(a) or (II)(g) (e.g., (II)(g)(iv)) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663 or known in the art. In some embodiments, the quantitation and/or sequencing of the extended capture probe and/or a denatured/separated single-stranded nucleic acid including a sequence that is complementary to the extended capture probe can be performed for about 10 minutes to about 5 hours (or any of the subranges of this range described herein).


Following the denaturing of the single-stranded nucleic acid including a sequence complementary to the extended capture probe, the single-stranded nucleic acid including the sequence complementary to the extended capture probe can be subjected to amplification, fragmentation, end-repairing, A-tailing, adaptor ligation, sample index PCR, and the construction and quality control of a gene expression library, using any of the exemplary methods described herein, or described in, e.g., Sections (II)(a) or (II)(g) (e.g., (II)(g)(iv)) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.



FIG. 3A is a diagram showing an exemplary reaction mix that can be used to generate a cDNA and that can be used to add (e.g., via extension) or ligate a second adaptor sequence to a 3′ end of the cDNA. For example, a target nucleic acid in the biological sample can be an mRNA molecule having a poly(A) tail at the 3′ end of the sequence. For the generation of a cDNA, a reverse transcription primer is added to the biological sample. The reverse transcription primer includes, from the 5′ end to the 3′ end, a first adaptor sequence (SM), and a sequence that is substantially complementary to a portion of the mRNA. In some embodiments, the sequence that is substantially complementary to a portion of the mRNA includes a poly(T) sequence comprising a sequence of Tn, wherein n can be 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30. In some embodiments, the sequence substantially complementary to a portion of the mRNA is a dT30VN sequence, where the sequence comprises Tn, wherein n is 30, wherein V is A, G, or C, and where N is A, G, C, or T. In some embodiments, the sequence substantially complementary to a portion of the mRNA can includes a random sequence.


In some embodiments, the complement of the second adaptor sequence can be added to a reverse transcription mix. In some embodiments, the complement of the second adaptor sequence can be a template switching oligonucleotide (TSO) having a rGrGrG sequence at the 3′ end of the second adaptor sequence.


The reverse transcription step can be performed using a method that includes a pre-equilibration thermocycling protocol (e.g., lid temperature and pre-equilibration at about 53° C., reverse transcription at about 53° C. for about 60 minutes, about 90° C. for about 5 min, and then held at about 4° C.). Any suitable reverse transcriptase and buffers can be used to perform reverse transcription, such as any of those described herein or known in the art. In some embodiments, the reverse transcription mix can further include other components that assist or increase the rate of a reverse transcription reaction. For example, the reverse transcription mix can further include dNTPs. The thermocycling protocol for the reverse transcription reaction and the ligation of the second adaptor sequence to the 3′ end of the cDNA can be further optimized according to different target nucleic acids in different types of biological samples.


The reaction described in FIG. 3A generates a cDNA comprising, for the 5′ end to the 3′ end, a first adaptor sequence (e.g., SM sequence), a sequence substantially complementary to the target nucleic acid (e.g., a dT30VN sequence), a sequence complementary to the target nucleic acid sequence, and a complement of a second adaptor sequence (e.g., a TSO sequence). In some embodiments, the cDNA is hybridized to the target nucleic acid molecule is subsequently denatured/separated from the nucleic acid analyte.



FIG. 3B is a diagram showing an exemplary array comprising a second exemplary capture probe. The capture probe comprises, from the 5′ end to the 3′ end, a linker sequence, a partial R1 primer sequence, a spatial barcode, a unique molecular identifier (UMI), a capture domain, e.g., a sequence substantially complementary to the second adaptor sequence. In some embodiments the sequence substantially complementary to the second adaptor sequence is substantially complementary to a template switching oligonucleotide (TSO) ligated to the cDNA molecule. In some embodiments the sequence substantially complementary to the second adaptor sequence is comprises a template switching oligonucleotide (TSO) used as a template for extension of a 3′ end of the cDNA molecule. In some embodiments, the 5′ end of the capture probe is attached to the array. After the capture domain on the capture probe specifically binds to the second adaptor sequence on the cDNA molecule, a 3′ end of the capture probe is extended (using the specifically bound cDNA as a template) to add a sequence that is substantially complementary to the sequence of the cDNA and a sequence complementary to the first adaptor sequence). In addition, a single-stranded nucleic acid that includes a sequence that is complementary to the extended capture probe can be generated (bottom strand shown in bottom half of figure).


The generation of the extended capture probe and the generation of the single-stranded nucleic acid that includes a sequence complementary to the extended capture probe in FIG. 3B can be performed using a thermocycling protocol (e.g., lid temperature and pre-equilibrate at about 95° C., denaturing at about 95° C. for about 1 min, reannealing at about 60° C. for about 60 min, extension at about 90° C. for about 5 minutes, and then held at about 4° C.). The reaction mixture further includes all necessary polymerase and buffers. In some embodiments, the polymerase can be a DNA polymerase. In some embodiments, the DNA polymerase can be HotStart Taq DNA polymerase.


After the generation of the single-stranded nucleic acid including a sequence that is complementary to the extended capture probe, KOH can be added to denature the single-stranded nucleic acid including a sequence complementary to the extended capture probe from the extended capture probe, and transferring the single-stranded nucleic acid including a sequence that is complementary to the extended capture probe to a different tube (e.g., one or more tubes, for example a strip tube that might be used in a thermocyling instrument) for the performance of additional steps.



FIG. 3C is a diagram showing exemplary steps of amplification, quantitation, and/or sequencing of a single-stranded nucleic acid that includes a sequence complementary to the extended capture probe. In some embodiments, the methods can include the performance of qPCR. Exemplary methods for performing qPCR are described herein and are known in the art.


In some embodiments, the method can result in the generation of a single-stranded nucleic acid that includes in a 5′ to a 3′ direction, a linker, a partial R1 primer sequence, a spatial barcode, a UMI, a sequence complementary to the second adaptor sequence, a sequence present in the target nucleic acid, and a sequence complementary to the first adaptor sequence.


In some embodiments, the method can result in the generation of a single-stranded nucleic acid that includes in a 5′ to a 3′ direction, a P5 sequencing handle, a i5 sequencing handle, a linker, a partial R1 or R1 primer sequence, a spatial barcode, a UMI, a sequence complementary to the second adaptor sequence, a sequence present in the target nucleic acid, a R2 adaptor sequence, an i7 sequencing handle, and a P7 sequencing handle.


In some embodiments, the method can result in the generation of a single-stranded nucleic acid that includes in a 3′ to a 5′ direction, a sequence complementary to a linker, a sequence complementary to a partial R1 primer sequence, a sequence complementary to a spatial barcode, a sequence complementary to a UMI, the second adaptor sequence, a sequence complementary to a sequence present in the target nucleic acid, and the first adaptor sequence.


In some embodiments, the method can result in the generation of a single-stranded nucleic acid that includes in a 3′ to a 5′ direction, a sequence complementary to a P5 sequencing handle, a sequence complementary to an i5 sequencing handle, a sequence complementary to a linker, a sequence complementary to a partial R1 or R1 primer sequence, a sequence complementary to a spatial barcode, a sequence complementary to a UMI, the second adaptor sequence, a sequence complementary to a sequence present in the target nucleic acid, a sequence complementary to an R2 adaptor sequence, a sequence complementary to an i7 sequencing handle, and a sequence complementary to a P7 sequencing handle.


In some embodiments of any of the methods described herein, step (e) includes sequencing all or a part of the sequence of the spatial barcode, or a complement thereof, and sequencing all of a part of the sequence of the target nucleic acid, or a complement thereof. The sequencing can be performed using any of the methods described herein. In some embodiments, step (e) includes sequencing the full-length sequence of the spatial barcode, or a complement thereof. In some embodiments, step (e) includes sequencing a part of the sequence of the spatial barcode, or a complement thereof. In some embodiments, step (e) includes sequencing the full-length sequence of the target nucleic acid, or a complement thereof. In some embodiments, step (e) includes sequencing a part of the target nucleic acid, or a complement thereof. In some embodiments, the sequencing is performed using high throughput sequencing. In some embodiments, the target nucleic acid is sequenced from the 5′ end of the target nucleic acid. In some embodiments, the target nucleic acid is sequenced from the 3′ end of the target nucleic acid. In some embodiments, the target nucleic acid is sequenced from both the 3′ end and the 5′ end of the target nucleic acid. The library can be sequenced using available sequencing platforms, including, any of MiSeq, NextSeq 500/550, HiSeq 2500, HiSeq 3000/4000, NovaSeq, or iSeq.


Kits


Also provided herein are kits for performing any of the methods described herein. For example, provided herein is a kit comprising: a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence; a reverse transcriptase; and an oligonucleotide comprising a second adaptor sequence or a complement thereof. In some embodiments, the reverse transcriptase is a reverse transcriptase with terminal transferase activity. In some embodiments, the second adaptor sequence or complement thereof is a TSO or complement thereof. The kits can include any other buffers, enzymes, cofactors, or other components useful in the method. For example, when the method includes ligating the second adaptor sequence to the generated cDNA molecule, the kit can further include a ligase. In some embodiments, the kits can also include an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence. In some examples, the kit can further include a permeabilizing agent. In some embodiments, the kit can further include a lipase, a protease, and/or an RNAse.


EXEMPLARY EMBODIMENTS

Embodiment 1 is a method of identifying a location of a target nucleic acid in a permeabilized biological sample, the method comprising:

    • (a) generating a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence, wherein the step of generating the cDNA molecule occurs within the permeabilized biological sample;
    • (b) ligating a second adaptor sequence to a 3′ end of the cDNA molecule, wherein the step of ligating is performed within the biological sample;
    • (c) releasing the cDNA molecule from the target nucleic acid, such that the cDNA contacts an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence ligated to the cDNA;
    • (d) extending a 3′ end of the capture probe using the cDNA as a template; and
    • (e) determining (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) all or a part of the sequence of the spatial barcode, or a complement thereof, and using the determined sequences of (i) and (ii) to identify the location of the target nucleic acid in the permeabilized biological sample.


Embodiment 2 is a method of identifying a location of a target nucleic acid in a permeabilized biological sample, the method comprising:

    • (a) generating a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence, wherein the step of generating the cDNA molecule occurs within the permeabilized biological sample;
    • (b) extending a 3′ end of the cDNA molecule to include a second adaptor sequence, wherein the step of extending is performed within the biological sample;
    • (c) releasing the cDNA molecule from the target nucleic acid, such that the cDNA contacts an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence;
    • (d) extending a 3′ end of the capture probe using the cDNA as a template; and
    • (e) determining (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) the sequence of the spatial barcode, or a complement thereof, and using the determined sequences of (i) and (ii) to identify the location of the target nucleic acid in the permeabilized biological sample.


Embodiment 3 is the method of Embodiment 2, wherein step (b) occurs simultaneously with step (a).


Embodiment 4 is the method of any one of Embodiments 1-3, wherein steps (a) through (c) are performed when the biological sample is disposed on the array.


Embodiment 5 is the method of any one of Embodiments 1-3, wherein step (a) is performed when the biological sample is not disposed on the array and step (b) is performed when the biological sample is disposed on the array, and wherein the method further comprises between steps (a) and (b), a step of disposing the biological sample on the array.


Embodiment 6 is the method of any one of Embodiments 1-3, wherein steps (a) and (b) are performed when the biological sample is not disposed on the array, and wherein the method further comprises between steps (b) and (c), a step of disposing the biological sample on the array.


Embodiment 7 is the method of any one of Embodiments 1-6, wherein the sequence that is substantially complementary to a portion of the target nucleic acid present in the reverse transcription primer comprises a poly(T) sequence.


Embodiment 8 is the method of any one of Embodiments 1-6, wherein the sequence that is substantially complementary to a portion of the target nucleic acid present in the reverse transcription primer comprises a random sequence.


Embodiment 9 is the method of any one of Embodiments 1-8, wherein the second adaptor sequence is a template switching oligonucleotide (TSO), or a complement thereof.


Embodiment 10 is the method of any one of Embodiments 1-9, wherein the array comprises a slide.


Embodiment 11 is the method of Embodiment 10, wherein a 5′ end of the capture probe is attached to the slide.


Embodiment 12 is the method of any one of Embodiments 1-9, wherein the array is a bead array.


Embodiment 13 is the method of Embodiment 12, wherein a 5′ end of the capture probe is attached to a bead of the bead array.


Embodiment 14 is the method of any one of Embodiments 1-13, wherein the capture probe further comprises a unique molecular identifier (UMI).


Embodiment 15 is the method of Embodiment 14, wherein the UMI is positioned 5′ relative to the capture domain in the capture probe.


Embodiment 16 is the method of any one of Embodiments 1-15, wherein the determining in step (e) comprises sequencing (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) the sequence of the spatial barcode, or a complement thereof.


Embodiment 17 is the method of Embodiment 16, wherein the sequencing is high throughput sequencing.


Embodiment 18 is the method of Embodiment 16, wherein the sequencing is sequencing by hybridization.


Embodiment 19 is the method of any one of Embodiments 1-18, wherein the target nucleic acid is RNA.


Embodiment 20 is the method of Embodiment 19, wherein the RNA is an mRNA.


Embodiment 21 is the method of any one of Embodiments 1-20, wherein the permeabilized biological sample is a permeabilized tissue section.


Embodiment 22 is the method of Embodiment 21, wherein the permeabilized tissue section is a permeabilized formalin-fixed and paraffin-embedded (FFPE) tissue section.


Embodiment 23 is the method of any one of Embodiments 1-22, wherein the method further comprises a step of imaging the biological sample.


Embodiment 24 is the method of Embodiment 23, wherein the step of imaging is performed prior to step (a).


Embodiment 25 is the method of Embodiment 24, wherein the step of imaging is performed between steps (b) and (c).


Embodiment 26 is the method of any one of Embodiments 1-3 and 6-25, wherein the method further comprises, between steps (b) and (c), a step of freezing and thawing the permeabilized biological sample.


Embodiment 27 is the method of Embodiment 26, wherein the method further comprises, between steps (b) and (c), a step of sectioning the permeabilized biological sample.


Embodiment 28 is the method of Embodiment 27, wherein the step of sectioning the permeabilized biological sample is performed using cryosectioning.


Embodiment 29 is the method of any one of Embodiments 1-28, wherein the method further comprises, prior to step (a), a step of permeabilizing the biological sample.


Embodiment 30 is the method of any one of Embodiments 1-29, wherein the performance of step (a) comprises introducing a reverse transcriptase, dNTPs, and the reverse transcription primer into the permeabilized biological sample.


Embodiment 31 is a kit comprising: a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence; a reverse transcriptase; and an oligonucleotide comprising a second adaptor sequence or a complement thereof.


Embodiment 32 is the kit of Embodiment 31, wherein the kit further comprises a ligase.


Embodiment 33 is the kit of Embodiment 30 or 31, wherein the reverse transcriptase is a reverse transcriptase with terminal transferase activity.


Embodiment 34 is the kit of any one of Embodiments 31-33, wherein the second adaptor sequence or the complement thereof is a TSO or a complement thereof.


Embodiment 35 is the kit of any one of Embodiments 31-34, wherein the kit further comprises an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds specifically to the second adaptor sequence.


Embodiment 36 is a nucleic acid comprising, in the 5′ to 3′ direction: a spatial barcode; a sequence complementary to a second adaptor sequence; a sequence present in a target nucleic acid; and a sequence complementary to a first adaptor sequence.


Embodiment 37 is a nucleic acid comprising, in the 3′ to 5′ direction: a complement of a spatial barcode; a second adaptor sequence; a sequence complementary to a sequence present in a target nucleic acid; and a first adaptor sequence.


Embodiment 38 is the nucleic acid of Embodiment 36 or 37, wherein the second adaptor sequence comprises a TSO.


Embodiment 39 is the nucleic acid of Embodiment 36 or 37, wherein the second adaptor sequence comprises a complement of a TSO.


Embodiment 40 is the nucleic acid of any one of Embodiments 36-39, wherein the first adaptor sequence is a reverse transcriptase primer.

Claims
  • 1. A method of identifying a location of a target nucleic acid in a permeabilized tissue section, the method comprising: (a) generating a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence, wherein the step of generating the cDNA molecule occurs within the permeabilized tissue section;(b) ligating a second adaptor sequence to a 3′ end of the cDNA molecule, wherein the step of ligating is performed within the tissue section;(c) releasing the cDNA molecule ligated to the second adaptor sequence from the target nucleic acid in the tissue section, such that the second adaptor sequence contacts an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that hybridizes to the second adaptor sequence ligated to the cDNA molecule;(d) extending a 3′ end of the capture probe using the cDNA molecule as a template; and(e) determining (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) the sequence of the spatial barcode, or a complement thereof, and using the determined sequences of (i) and (ii) to identify the location of the target nucleic acid in the permeabilized tissue section.
  • 2. A method of identifying a location of a target nucleic acid in a permeabilized tissue section, the method comprising: (a) generating a cDNA molecule comprising a sequence that is substantially complementary to the target nucleic acid using a reverse transcription primer comprising (i) a sequence that is substantially complementary to a portion of the target nucleic acid and (ii) a first adaptor sequence, wherein the step of generating the cDNA molecule occurs within the permeabilized tissue section;(b) extending a 3′ end of the cDNA molecule to include a second adaptor sequence thereby generating an extended cDNA molecule, wherein the step of extending is performed within the tissue section;(c) releasing the extended cDNA molecule from the target nucleic acid, such that the extended cDNA molecule contacts an array, wherein the array comprises an attached capture probe comprising in a 5′ to a 3′ direction: (i) a spatial barcode and (ii) a capture domain that binds to the second adaptor sequence of the extended cDNA molecule;(d) extending a 3′ end of the capture probe using the extended cDNA molecule as a template; and(e) determining (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) the sequence of the spatial barcode, or a complement thereof, and using the determined sequences of (i) and (ii) to identify the location of the target nucleic acid in the permeabilized tissue section.
  • 3. The method claim 1, wherein steps (a) through (c) are performed when the tissue section is disposed on the array.
  • 4. The method of claim 1, wherein step (a) is performed when the tissue section is not disposed on the array and step (b) is performed when the tissue section is disposed on the array, and wherein the method further comprises between steps (a) and (b), a step of disposing the tissue section on the array.
  • 5. The method of claim 1, wherein steps (a) and (b) are performed when the tissue section is not disposed on the array, and wherein the method further comprises between steps (b) and (c), a step of disposing the tissue section on the array.
  • 6. The method of claim 1, wherein the sequence that is substantially complementary to a portion of the target nucleic acid present in the reverse transcription primer comprises a poly(T) sequence.
  • 7. The method of claim 1, wherein the sequence that is substantially complementary to a portion of the target nucleic acid present in the reverse transcription primer comprises a random sequence.
  • 8. The method of claim 1, wherein the second adaptor sequence is a template switching oligonucleotide (TSO), or a complement thereof.
  • 9. The method of claim 1, wherein the capture probe further comprises a unique molecular identifier (UMI).
  • 10. The method of claim 1, wherein the determining in step (e) comprises sequencing (i) all or a part of the sequence of the target nucleic acid, or a complement thereof, and (ii) the sequence of the spatial barcode, or a complement thereof.
  • 11. The method of claim 10, wherein the sequencing is high throughput sequencing.
  • 12. The method of claim 10, wherein the sequencing is sequencing by hybridization.
  • 13. The method of claim 1, wherein the target nucleic acid is RNA.
  • 14. The method of claim 13, wherein the RNA is an mRNA.
  • 15. The method of claim 1, wherein the method further comprises, prior to step (a), a step of permeabilizing the tissue section.
  • 16. The method of claim 1, wherein step (a) comprises introducing a reverse transcriptase, dNTPs, and the reverse transcription primer into the permeabilized tissue section.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority to U.S. Provisional Patent Application Ser. No. 62/980,867, filed Feb. 24, 2020; the entire contents of which are herein incorporated by reference.

US Referenced Citations (774)
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
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
5695940 Drmanac et al. Dec 1997 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
5925545 Reznikoff et al. Jul 1999 A
5928906 Koester et al. Jul 1999 A
5958775 Wickstrrom Sep 1999 A
5965443 Reznikoff et al. Oct 1999 A
6013440 Lipshutz Jan 2000 A
6027889 Barany et al. Feb 2000 A
6054274 Sampson et al. Apr 2000 A
6060240 Kamb et al. May 2000 A
6130073 Eggerding Oct 2000 A
6143496 Brown Nov 2000 A
6153389 Haarer Nov 2000 A
6159736 Reznikoff et al. Dec 2000 A
6165714 Lane et al. Dec 2000 A
6210891 Nyren Apr 2001 B1
6210894 Brennan Apr 2001 B1
6214587 Dattagupta Apr 2001 B1
6251639 Kurn Jun 2001 B1
6258568 Nyren Jul 2001 B1
6266459 Walt Jul 2001 B1
6268148 Barany et al. Jul 2001 B1
6274320 Rothberg Aug 2001 B1
6291180 Chu Sep 2001 B1
6291187 Kingsmore et al. Sep 2001 B1
6300063 Lipshutz et al. Oct 2001 B1
6309824 Drmanac Oct 2001 B1
6323009 Lasken et al. Nov 2001 B1
6344316 Lockhart Feb 2002 B1
6344329 Lizardi et al. 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
6534266 Singer Mar 2003 B1
6544732 Chee Apr 2003 B1
6573043 Cohen et al. Jun 2003 B1
6579695 Lambalot Jun 2003 B1
6620584 Chee Sep 2003 B1
6632641 Brennan Oct 2003 B1
6737236 Pieken et al. May 2004 B1
6770441 Dickinson Aug 2004 B2
6773886 Kaufman Aug 2004 B2
6787308 Balasubramanian Sep 2004 B2
6797470 Barany et al. 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
6852487 Barany et al. Feb 2005 B1
6859570 Walt Feb 2005 B2
6864052 Drmanac Mar 2005 B1
6867028 Janulaitis Mar 2005 B2
6872816 Hall et al. Mar 2005 B1
6875572 Prudent et al. Apr 2005 B2
6890741 Fan et al. May 2005 B2
6897023 Fu May 2005 B2
6913881 Aizenstein et al. Jul 2005 B1
6942968 Dickinson et al. Sep 2005 B1
7011944 Prudent et al. Mar 2006 B2
7057026 Barnes Jun 2006 B2
7083980 Reznikoff et al. Aug 2006 B2
7115400 Adessi Oct 2006 B1
7118883 Inoue Oct 2006 B2
7166431 Chee et al. Jan 2007 B2
7192735 Lambalot Mar 2007 B2
7211414 Hardin May 2007 B2
7255994 Lao Aug 2007 B2
7258976 Mitsuhashi Aug 2007 B2
7282328 Kong et al. Oct 2007 B2
7297518 Quake Nov 2007 B2
7329492 Hardin Feb 2008 B2
7358047 Hafner et al. Apr 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
7473767 Dimitrov Jan 2009 B2
7537897 Brenner May 2009 B2
7563576 Chee Jul 2009 B2
7579153 Brenner Aug 2009 B2
7582420 Oliphant et al. Sep 2009 B2
7601498 Mao Oct 2009 B2
7608434 Reznikoff et al. Oct 2009 B2
7611869 Fan Nov 2009 B2
7635566 Brenner Dec 2009 B2
7666612 Johnsson Feb 2010 B2
7674752 He Mar 2010 B2
7709198 Luo et al. May 2010 B2
7776547 Roth Aug 2010 B2
7776567 Mao Aug 2010 B2
7803943 Mao Sep 2010 B2
7888009 Barany et al. Feb 2011 B2
7892747 Barany et al. Feb 2011 B2
7910304 Drmanac Mar 2011 B2
7914981 Barany et al. Mar 2011 B2
7955794 Shen et al. Jun 2011 B2
7960119 Chee Jun 2011 B2
7985565 Mayer et al. Jul 2011 B2
8003354 Shen et al. Aug 2011 B2
8076063 Fan Dec 2011 B2
8092784 Mao Jan 2012 B2
8148068 Brenner Apr 2012 B2
8206917 Chee Jun 2012 B2
8268554 Schallmeiner Sep 2012 B2
8288103 Oliphant Oct 2012 B2
8288122 O'Leary et al. Oct 2012 B2
8383338 Kitzman Feb 2013 B2
8431691 McKernan et al. Apr 2013 B2
8460865 Chee Jun 2013 B2
8481257 Van Eijk Jul 2013 B2
8481258 Church et al. Jul 2013 B2
8481292 Casbon Jul 2013 B2
8481698 Lieberman et al. Jul 2013 B2
8507204 Pierce et al. Aug 2013 B2
8519115 Webster et al. Aug 2013 B2
8551710 Bernitz et al. Oct 2013 B2
8568979 Stuelpnagel et al. Oct 2013 B2
RE44596 Stroun et al. Nov 2013 E
8586310 Mitra Nov 2013 B2
8597891 Barany et al. Dec 2013 B2
8603743 Liu et al. Dec 2013 B2
8604182 Luo et al. Dec 2013 B2
8614073 Van Eijk Dec 2013 B2
8624016 Barany et al. Jan 2014 B2
8685889 Van Eijk Apr 2014 B2
8741564 Seligmann Jun 2014 B2
8741606 Casbon Jun 2014 B2
8771950 Church et al. Jul 2014 B2
8785353 Van Eijk Jul 2014 B2
8790873 Namsaraev et al. Jul 2014 B2
8809238 Livak et al. Aug 2014 B2
8815512 Van Eijk Aug 2014 B2
8835358 Fodor Sep 2014 B2
8865410 Shendure Oct 2014 B2
8906626 Oliphant et al. Dec 2014 B2
8911945 Van Eijk Dec 2014 B2
8936912 Mitra Jan 2015 B2
8951726 Luo et al. Feb 2015 B2
8951728 Rasmussen Feb 2015 B2
8986926 Ferree et al. Mar 2015 B2
9005891 Sinicropi et al. Apr 2015 B2
9023768 Van Eijk May 2015 B2
9062348 Van Eijk Jun 2015 B1
9080210 Van Eijk Jul 2015 B2
9194001 Brenner Nov 2015 B2
9201063 Sood et al. Dec 2015 B2
9273349 Nguyen et al. Mar 2016 B2
9290808 Fodor Mar 2016 B2
9290809 Fodor Mar 2016 B2
9328383 Van Eijk May 2016 B2
9334536 Van Eijk May 2016 B2
9371563 Geiss et al. Jun 2016 B2
9371598 Chee Jun 2016 B2
9376716 Van Eijk Jun 2016 B2
9376717 Gao et al. Jun 2016 B2
9376719 Van Eijk Jun 2016 B2
9416409 Hayden Aug 2016 B2
9447459 Van Eijk Sep 2016 B2
9453256 Van Eijk Sep 2016 B2
9493820 Van Eijk Nov 2016 B2
9506061 Brown Nov 2016 B2
9512422 Barnard et al. Dec 2016 B2
9574230 Van Eijk Feb 2017 B2
9593365 Frisen et al. Mar 2017 B2
9598728 Barany et al. Mar 2017 B2
9624538 Church et al. Apr 2017 B2
9644204 Hindson et al. May 2017 B2
9657335 Van Eijk May 2017 B2
9670542 Van Eijk Jun 2017 B2
9694361 Bharadwaj Jul 2017 B2
9702004 Van Eijk Jul 2017 B2
9714446 Webster et al. Jul 2017 B2
9714937 Dunaway Jul 2017 B2
9727810 Fodor et al. Aug 2017 B2
9745627 Van Eijk Aug 2017 B2
9777324 Van Eijk Oct 2017 B2
9783841 Nolan et al. Oct 2017 B2
9816134 Namsaraev Nov 2017 B2
9834814 Peter et al. Dec 2017 B2
9850536 Oliphant et al. Dec 2017 B2
9856521 Stevens et al. Jan 2018 B2
9868979 Chee et al. Jan 2018 B2
9879313 Chee et al. Jan 2018 B2
9896721 Van Eijk Feb 2018 B2
9898576 Van Eijk Feb 2018 B2
9898577 Van Eijk Feb 2018 B2
9902991 Sinicropi et al. Feb 2018 B2
9909167 Samusik et al. Mar 2018 B2
9938566 Shepard et al. Apr 2018 B2
9957550 Yeakley et al. May 2018 B2
10002316 Fodor et al. Jun 2018 B2
10023907 Van Eijk Jul 2018 B2
10030261 Frisen et al. Jul 2018 B2
10035992 Gloeckner et al. Jul 2018 B2
10041949 Bendall et al. Aug 2018 B2
10059989 Giresi et al. Aug 2018 B2
10059990 Boyden et al. Aug 2018 B2
10095832 Van Eijk Oct 2018 B2
10144966 Cantor Dec 2018 B2
10208982 Bannish et al. Feb 2019 B2
10227639 Levner et al. Mar 2019 B2
10273541 Hindson et al. Apr 2019 B2
10357771 Bharadwaj Jul 2019 B2
10370698 Nolan et al. Aug 2019 B2
10415080 Dunaway et al. Sep 2019 B2
10465235 Gullberg et al. Nov 2019 B2
10472669 Chee Nov 2019 B2
10480022 Chee Nov 2019 B2
10480029 Bent et al. Nov 2019 B2
10494667 Chee Dec 2019 B2
10495554 Deisseroth et al. Dec 2019 B2
10501777 Beechem et al. Dec 2019 B2
10501791 Church et al. Dec 2019 B2
10510435 Cai et al. Dec 2019 B2
10544403 Gloeckner et al. Jan 2020 B2
10550429 Harada et al. Feb 2020 B2
10590244 Delaney et al. Mar 2020 B2
10633648 Seelig et al. Apr 2020 B2
10640816 Beechem et al. May 2020 B2
10640826 Church et al. May 2020 B2
10669569 Gullberg et al. Jun 2020 B2
10724078 Van Driel et al. Jul 2020 B2
10725027 Bell Jul 2020 B2
10774372 Chee et al. Sep 2020 B2
10774374 Frisen et al. Sep 2020 B2
10787701 Chee Sep 2020 B2
10815519 Husain et al. Oct 2020 B2
10829803 Terbrueggen et al. Nov 2020 B2
10844426 Daugharthy et al. Nov 2020 B2
10858698 Church et al. Dec 2020 B2
10858702 Lucero et al. Dec 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
11104936 Zhang et al. Aug 2021 B2
11118216 Koshinsky et al. Sep 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
11352667 Hauling et al. Jun 2022 B2
11359228 Chee et al. Jun 2022 B2
11365442 Chee Jun 2022 B2
11371086 Chee Jun 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 et al. Dec 2022 B2
11530438 Persson et al. Dec 2022 B2
11535887 Gallant et al. Dec 2022 B2
11542543 Chee Jan 2023 B2
11549138 Chee Jan 2023 B2
11560587 Chee Jan 2023 B2
11560592 Chew et al. Jan 2023 B2
11560593 Chell et al. Jan 2023 B2
11592447 Uytingco et al. Feb 2023 B2
11608498 Gallant et al. Mar 2023 B2
11608520 Galonska et al. Mar 2023 B2
11613773 Frisen et al. Mar 2023 B2
11618897 Kim et al. Apr 2023 B2
11618918 Chee et al. Apr 2023 B2
11624063 Dadhwal Apr 2023 B2
11624086 Uytingco et al. Apr 2023 B2
11634756 Chee Apr 2023 B2
11611626 Katiraee et al. May 2023 B1
11649485 Yin et al. May 2023 B2
11661626 Katiraee et al. May 2023 B2
11680260 Kim et al. Jun 2023 B2
11692218 Engblom et al. Jul 2023 B2
11702693 Bharadwaj Jul 2023 B2
11702698 Stoeckius Jul 2023 B2
11732292 Chee Aug 2023 B2
11732299 Ramachandran Iyer Aug 2023 B2
11732300 Bava Aug 2023 B2
11733238 Chee Aug 2023 B2
11739372 Frisen et al. Aug 2023 B2
11739381 Chew et al. Aug 2023 B2
11753673 Chew et al. Sep 2023 B2
11753674 Chee et al. Sep 2023 B2
11753675 Ramachandran Iyer Sep 2023 B2
11761030 Chee Sep 2023 B2
11761038 Stoeckius Sep 2023 B1
11767550 Chee Sep 2023 B2
11768175 Kim et al. Sep 2023 B1
11773433 Gallant et al. Oct 2023 B2
11781130 Dadhwal Oct 2023 B2
11788122 Frisen et al. Oct 2023 B2
11795498 Frisen et al. Oct 2023 B2
11795507 Chell et al. Oct 2023 B2
11808769 Uytingco et al. Nov 2023 B2
11821024 Chee et al. Nov 2023 B2
11821035 Bent et al. Nov 2023 B1
11827935 Ramachandran Iyer et al. Nov 2023 B1
11835462 Bava Dec 2023 B2
11840687 Gallant et al. Dec 2023 B2
11840724 Chew et al. Dec 2023 B2
20010055764 Empendocles et al. Dec 2001 A1
20020040275 Cravatt Apr 2002 A1
20020051986 Baez et al. May 2002 A1
20020055100 Kawashima May 2002 A1
20020086441 Baranov et al. Jul 2002 A1
20020164611 Bamdad Nov 2002 A1
20030017451 Wang et al. Jan 2003 A1
20030022207 Balasubramanian Jan 2003 A1
20030064398 Barnes Apr 2003 A1
20030138879 Lambalot Jul 2003 A1
20030148335 Shen et al. Aug 2003 A1
20030162216 Gold Aug 2003 A1
20030165948 Alsmadi et al. Sep 2003 A1
20030211489 Shen et al. Nov 2003 A1
20030224419 Corcoran Dec 2003 A1
20030232348 Jones et al. Dec 2003 A1
20030232382 Brennan Dec 2003 A1
20030235854 Chan et al. Dec 2003 A1
20040033499 Ilsley et al. Feb 2004 A1
20040067492 Peng et al. Apr 2004 A1
20040082059 Webb et al. Apr 2004 A1
20040096853 Mayer May 2004 A1
20040106110 Balasubramanian Jun 2004 A1
20040235103 Reznikoff et al. Nov 2004 A1
20040248325 Bukusoglu et al. Dec 2004 A1
20040259105 Fan et al. Dec 2004 A1
20050003431 Wucherpfennig Jan 2005 A1
20050014203 Darfler et al. Jan 2005 A1
20050037393 Gunderson et al. Feb 2005 A1
20050048580 Labaer Mar 2005 A1
20050064460 Holliger et al. Mar 2005 A1
20050095627 Kolman et al. May 2005 A1
20050100900 Kawashima et al. May 2005 A1
20050130173 Leamon et al. Jun 2005 A1
20050136414 Gunderson et al. Jun 2005 A1
20050164292 Farooqui Jul 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
20050239119 Tsukada et al. Oct 2005 A1
20050260653 LaBaer Nov 2005 A1
20050266417 Barany et al. Dec 2005 A1
20060046313 Roth Mar 2006 A1
20060084078 Zhao Apr 2006 A1
20060105352 Qiao et al. May 2006 A1
20060154286 Kong et al. Jul 2006 A1
20060188901 Barnes et al. Aug 2006 A1
20060199183 Valat et al. Sep 2006 A1
20060211001 Yu et al. Sep 2006 A1
20060216775 Burkart et al. Sep 2006 A1
20060240439 Smith et al. Oct 2006 A1
20060263789 Kincaid Nov 2006 A1
20060275782 Gunderson et al. Dec 2006 A1
20060281109 Barr Ost et al. Dec 2006 A1
20070020640 McCloskey et al. Jan 2007 A1
20070020669 Ericsson Jan 2007 A1
20070026430 Andersen et al. Feb 2007 A1
20070054288 Su et al. Mar 2007 A1
20070087360 Boyd Apr 2007 A1
20070099208 Drmanac et al. May 2007 A1
20070128624 Gormley et al. Jun 2007 A1
20070128656 Agrawal Jun 2007 A1
20070134723 Kozlov et al. Jun 2007 A1
20070161020 Luo et al. Jul 2007 A1
20070166705 Milton et al. Jul 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
20080003586 Hyde et al. Jan 2008 A1
20080009420 Schroth et al. Jan 2008 A1
20080108082 Rank et al. May 2008 A1
20080108804 Hayashizaki et al. May 2008 A1
20080132429 Perov 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
20080293046 Allawi 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
20090036323 van Eijk et al. Feb 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
20090170713 van Eijk et al. Jul 2009 A1
20090202998 Schlumpberger et al. Aug 2009 A1
20090233802 Bignell et al. Sep 2009 A1
20090253581 van Eijk et al. Oct 2009 A1
20090283407 Van Eijk Nov 2009 A1
20090291854 Weisinger-Mayr et al. Nov 2009 A1
20090312193 Kim et al. Dec 2009 A1
20100035249 Hayashizaki et al. Feb 2010 A1
20100069263 Shendure et al. Mar 2010 A1
20100105052 Drmanac et al. Apr 2010 A1
20100120097 Matz et al. May 2010 A1
20100120098 Grunenwald et al. May 2010 A1
20100129874 Mitra et al. May 2010 A1
20100145037 Brive et al. Jun 2010 A1
20100173384 Johnsson et al. Jul 2010 A1
20100184618 Namsaraev et al. Jul 2010 A1
20100210475 Lee et al. Aug 2010 A1
20100227329 Cuppens Sep 2010 A1
20100273219 May et al. Oct 2010 A1
20110028685 Purkayastha et al. Feb 2011 A1
20110033854 Drmanac et al. Feb 2011 A1
20110045462 Fu et al. Feb 2011 A1
20110059436 Hardin et al. Mar 2011 A1
20110111409 Sinicropi et al. May 2011 A1
20110152111 Illumina Jun 2011 A1
20110245101 Chee et al. Oct 2011 A1
20110245111 Chee Oct 2011 A1
20110287435 Grunenwald et al. Nov 2011 A1
20120046175 Rodesch et al. Feb 2012 A1
20120046178 Van Den Boom et al. Feb 2012 A1
20120065081 Chee Mar 2012 A1
20120135871 van Eijk et al. May 2012 A1
20120202698 van Eijk et al. Aug 2012 A1
20120202704 Fan et al. Aug 2012 A1
20120245053 Shirai et al. Sep 2012 A1
20120252702 Muratani et al. Oct 2012 A1
20120258871 Kozlov et al. Oct 2012 A1
20120289414 Mitra et al. Nov 2012 A1
20130005594 Terbrueggen et al. Jan 2013 A1
20130005600 Olek Jan 2013 A1
20130023433 Luo et al. Jan 2013 A1
20130035239 Kong et al. Feb 2013 A1
20130079232 Kain et al. Mar 2013 A1
20130171621 Luo et al. Jul 2013 A1
20130244884 Jacobson et al. Sep 2013 A1
20130261019 Lin et al. Oct 2013 A1
20130338042 Shen et al. Dec 2013 A1
20140066318 Frisen et al. Mar 2014 A1
20140121118 Warner May 2014 A1
20140270435 Dunn Sep 2014 A1
20140274731 Raymond et al. Sep 2014 A1
20140323330 Glezer et al. Oct 2014 A1
20140342921 Weiner Nov 2014 A1
20140378350 Hindson et al. Dec 2014 A1
20150000854 Gann-Fetter et al. Jan 2015 A1
20150292988 Bharadwaj et al. Oct 2015 A1
20150344942 Frisen et al. Dec 2015 A1
20160019337 Roberts et al. Jan 2016 A1
20160024576 Chee Jan 2016 A1
20160041159 Labaer et al. Feb 2016 A1
20160060687 Zhu et al. Mar 2016 A1
20160108458 Frei et al. Apr 2016 A1
20160122817 Jarosz et al. May 2016 A1
20160138091 Chee et al. May 2016 A1
20160145677 Chee et al. May 2016 A1
20160201125 Samuels et al. Jul 2016 A1
20160253584 Fodor et al. Sep 2016 A1
20160289740 Fu et al. Oct 2016 A1
20160298180 Chee Oct 2016 A1
20160305856 Boyden et al. Oct 2016 A1
20160333403 Chee Nov 2016 A1
20160376642 Landegren et al. Dec 2016 A1
20170009278 Söderberg et al. Jan 2017 A1
20170016053 Beechem et al. Jan 2017 A1
20170029875 Zhang et al. Feb 2017 A1
20170058339 Chee Mar 2017 A1
20170058340 Chee Mar 2017 A1
20170058345 Chee Mar 2017 A1
20170067096 Wassie et al. Mar 2017 A1
20170088881 Chee Mar 2017 A1
20170089811 Tillberg et al. Mar 2017 A1
20170283860 Kool et al. Apr 2017 A1
20170166962 van Eijk et al. Jun 2017 A1
20170220733 Zhuang et al. Aug 2017 A1
20170233722 Seelig et al. Aug 2017 A1
20170241911 Rockel et al. Aug 2017 A1
20170335297 Ha et al. Nov 2017 A1
20170335410 Driscoll et al. Nov 2017 A1
20170342405 Fu et al. Nov 2017 A1
20170349940 Morin et al. Dec 2017 A1
20180051322 Church et al. Feb 2018 A1
20180057873 Zhou et al. Mar 2018 A1
20180080019 Blainey et al. Mar 2018 A1
20180094316 Oliphant et al. Apr 2018 A1
20180112261 Van Driel et al. Apr 2018 A1
20180127817 Borchert et al. May 2018 A1
20180163265 Zhang et al. Jun 2018 A1
20180179591 van Eijk Jun 2018 A1
20180201925 Steemers et al. Jul 2018 A1
20180201980 Chee et al. Jul 2018 A1
20180208967 Larman et al. Jul 2018 A1
20180216161 Chen et al. Aug 2018 A1
20180216162 Belhocine et al. Aug 2018 A1
20180237864 Imler et al. Aug 2018 A1
20180245142 So et al. Aug 2018 A1
20180247017 van Eijk et al. Aug 2018 A1
20180291427 Edelman Oct 2018 A1
20180291439 van Eijk et al. Oct 2018 A1
20180305681 Jovanovich et al. Oct 2018 A1
20180312822 Lee et al. Nov 2018 A1
20180320226 Church et al. Nov 2018 A1
20190055594 Samusik et al. Feb 2019 A1
20190064173 Bharadwaj et al. Feb 2019 A1
20190071656 Chang et al. Mar 2019 A1
20190085383 Church et al. Mar 2019 A1
20190119735 Deisseroth et al. Apr 2019 A1
20190135774 Orbai May 2019 A1
20190145982 Chee et al. May 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
20190177800 Boutet et al. Jun 2019 A1
20190194709 Church et al. Jun 2019 A1
20190203275 Frisen et al. Jul 2019 A1
20190218276 Regev et al. Jul 2019 A1
20190218608 Daugharthy 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
20190271028 Khafizov et al. Sep 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
20190360034 Zhou et al. Nov 2019 A1
20190367969 Belhocine et al. Dec 2019 A1
20190367982 Belhocine et al. Dec 2019 A1
20190367997 Bent et al. Dec 2019 A1
20200002763 Belgrader et al. Jan 2020 A1
20200010891 Beechem et al. Jan 2020 A1
20200024641 Nolan et al. Jan 2020 A1
20200047010 Lee et al. Feb 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
20200123597 Daniel Apr 2020 A1
20200140920 Pierce et al. May 2020 A1
20200173985 Dong et al. Jun 2020 A1
20200199565 Chen et al. Jun 2020 A1
20200199572 Kuersten et al. Jun 2020 A1
20200224244 Nilsson et al. Jul 2020 A1
20200239874 Mikkelsen 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
20200283852 Oliphant et al. Sep 2020 A1
20200299757 Chee et al. Sep 2020 A1
20200325531 Chee Oct 2020 A1
20200362398 Kishi 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
20210017587 Cai et al. Jan 2021 A1
20210095331 Fan et al. Apr 2021 A1
20210115504 Cai et al. Apr 2021 A1
20210123040 Macosko et al. Apr 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
20210238581 Mikkelsen et al. Aug 2021 A1
20210238664 Bava et al. Aug 2021 A1
20210238675 Bava et al. 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
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
20220325325 Chee et al. Oct 2022 A1
20220326251 Uytingco et al. Oct 2022 A1
20220333171 Chee Oct 2022 A1
20220333191 Mikkelsen et al. 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
20220389503 Mikkelsen et al. 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
20230033960 Gallant et al. Feb 2023 A1
20230034039 Shahjamali Feb 2023 A1
20230034216 Bava Feb 2023 A1
20230040363 Chee Feb 2023 A1
20230042088 Chee Feb 2023 A1
20230042817 Mignardi Feb 2023 A1
20230047782 Tentori et al. Feb 2023 A1
20230056549 Dadhwal Feb 2023 A1
20230064372 Chell et al. Mar 2023 A1
20230069046 Chew et al. Mar 2023 A1
20230077364 Patterson et al. Mar 2023 A1
20230080543 Katiraee et al. Mar 2023 A1
20230081381 Chew et al. Mar 2023 A1
20230100497 Frisen et al. Mar 2023 A1
20230107023 Chee Apr 2023 A1
20230111225 Chew et al. Apr 2023 A1
20230113230 Kim et al. Apr 2023 A1
20230126825 Nagendran et al. Apr 2023 A1
20230129552 Ramachandran Iyer Apr 2023 A1
20230135010 Tentori et al. May 2023 A1
20230143569 Iyer et al. May 2023 A1
20230145575 Gallant et al. May 2023 A1
20230147726 Hadrup et al. May 2023 A1
20230151412 Chee May 2023 A1
20230159994 Chee May 2023 A1
20230159995 Iyer et al. May 2023 A1
20230160008 Chell et al. May 2023 A1
20230175045 Katsori et al. Jun 2023 A1
20230183785 Frisen et al. Jun 2023 A1
20230194469 Tentori et al. Jun 2023 A1
20230194470 Kim et al. Jun 2023 A1
20230203478 Kim et al. Jun 2023 A1
20230183684 Gallant et al. Jul 2023 A1
20230212650 Chew et al. Jul 2023 A1
20230212655 Chee Jul 2023 A1
20230220368 Kim Jul 2023 A1
20230220454 Bent et al. Jul 2023 A1
20230220455 Galonska et al. Jul 2023 A1
20230227811 Dadhwal Jul 2023 A1
20230228762 Uytingco et al. Jul 2023 A1
20230242973 Frisen et al. Aug 2023 A1
20230242976 Tentori et al. Aug 2023 A1
20230265488 Gohil et al. Aug 2023 A1
20230265489 Uytingco et al. Aug 2023 A1
20230265491 Tentori et al. Aug 2023 A1
20230279474 Katiraee Sep 2023 A1
20230279477 Kvastad et al. Sep 2023 A1
20230279481 Marrache et al. Sep 2023 A1
20230287399 Gallant et al. Sep 2023 A1
20230287475 Chell et al. Sep 2023 A1
20230287481 Katsori et al. Sep 2023 A1
20230295699 Sukovich et al. Sep 2023 A1
20230295722 Bharadwaj Sep 2023 A1
20230304074 Chee et al. Sep 2023 A1
20230304078 Frisen et al. Sep 2023 A1
20230313279 Giacomello et al. Oct 2023 A1
20230323340 Dadhwal Oct 2023 A1
20230323434 Yin et al. Oct 2023 A1
20230323436 Chee Oct 2023 A1
20230323447 Schnall-Levin et al. Oct 2023 A1
20230323453 Stoeckius Oct 2023 A1
20230332138 Kim et al. Oct 2023 A1
20230332211 Chee Oct 2023 A1
20230332212 Chew et al. Oct 2023 A1
20230332227 Ramachandran Iyer Oct 2023 A1
20230332247 Singh et al. Oct 2023 A1
20230358733 Chee Nov 2023 A1
20230366008 Chew et al. Nov 2023 A1
20230383285 Kim et al. Nov 2023 A1
20230383344 Stoeckius Nov 2023 A1
Foreign Referenced Citations (211)
Number Date Country
2003200718 Oct 2006 AU
1273609 Nov 2000 CN
1537953 Oct 2004 CN
1680604 Oct 2005 CN
1749752 Mar 2006 CN
1898398 Jan 2007 CN
101142325 Mar 2008 CN
101221182 Jul 2008 CN
101522915 Sep 2009 CN
108949924 Dec 2018 CN
1782737 May 2007 EP
1910562 Apr 2008 EP
1923471 May 2008 EP
2002017 Dec 2008 EP
2292788 Mar 2011 EP
2302070 Mar 2011 EP
2881465 Jun 2015 EP
3013984 May 2016 EP
3511423 Jul 2019 EP
3541956 Sep 2019 EP
2520765 Jun 2015 GB
2007-014297 Jan 2007 JP
2007-074967 Mar 2007 JP
2009-036694 Feb 2009 JP
WO 1989010977 Nov 1989 WO
WO 1991006678 May 1991 WO
WO 1993004199 Mar 1993 WO
WO 1995023875 Sep 1995 WO
WO 1995025116 Sep 1995 WO
WO 1995035505 Dec 1995 WO
WO 1997031256 Aug 1997 WO
WO 200017390 Mar 2000 WO
WO 200106012 Jan 2001 WO
WO 2001009363 Feb 2001 WO
WO 2001012862 Feb 2001 WO
WO 2001042796 Jun 2001 WO
WO 2001046402 Jun 2001 WO
WO 2001090415 Nov 2001 WO
WO 2001096608 Dec 2001 WO
WO 2002059355 Aug 2002 WO
WO 2002059364 Aug 2002 WO
WO 2002077283 Oct 2002 WO
WO 2003002979 Jan 2003 WO
WO 2003008538 Jan 2003 WO
WO 2003010176 Feb 2003 WO
WO 2003102233 Dec 2003 WO
WO 2004015080 Feb 2004 WO
WO 2004067759 Aug 2004 WO
WO 2004081225 Sep 2004 WO
WO 2005007814 Jan 2005 WO
WO 2005010145 Feb 2005 WO
WO 2005026387 Mar 2005 WO
WO 2005042759 May 2005 WO
WO 2005113804 Dec 2005 WO
WO 2006020515 Feb 2006 WO
WO 2006124771 Nov 2006 WO
WO 2007041689 Apr 2007 WO
WO 2007060599 May 2007 WO
WO 2007073171 Jun 2007 WO
WO 2007076726 Jul 2007 WO
WO 2007139766 Dec 2007 WO
WO 2007145612 Dec 2007 WO
WO 2008069906 Jun 2008 WO
WO 2009032167 Mar 2009 WO
WO 2009152928 Dec 2009 WO
WO 2010019826 Feb 2010 WO
WO 2010027870 Mar 2010 WO
WO 2010126614 Nov 2010 WO
WO 2011008502 Jan 2011 WO
WO 2011062933 May 2011 WO
WO 2011068088 Jun 2011 WO
WO 2012049316 Apr 2012 WO
WO 2012061832 May 2012 WO
WO 2012071428 May 2012 WO
WO 2012129242 Sep 2012 WO
WO 2012159089 Nov 2012 WO
WO 2013123442 Aug 2013 WO
WO 2013131962 Sep 2013 WO
WO 2013138510 Sep 2013 WO
WO 2013142389 Sep 2013 WO
WO 2013150083 Oct 2013 WO
WO 2014044724 Mar 2014 WO
WO 2014060483 Apr 2014 WO
WO 2014071361 May 2014 WO
WO 2014144713 Sep 2014 WO
WO 2014152397 Sep 2014 WO
WO 2014210223 Dec 2014 WO
WO 2014210225 Dec 2014 WO
WO 2014210353 Dec 2014 WO
WO 2015031691 Mar 2015 WO
WO 2015161173 Oct 2015 WO
WO 2016077763 May 2016 WO
WO 2016138496 Sep 2016 WO
WO 2016138500 Sep 2016 WO
WO 2016166128 Oct 2016 WO
WO 2016168825 Oct 2016 WO
WO 2016172362 Oct 2016 WO
WO 2017019456 Feb 2017 WO
WO 2017019481 Feb 2017 WO
WO 2017075293 May 2017 WO
WO 2017096158 Jul 2017 WO
WO 2017143155 Aug 2017 WO
WO 2017156336 Sep 2017 WO
WO 2017184984 Oct 2017 WO
WO 2017192633 Nov 2017 WO
WO 2018023068 Feb 2018 WO
WO 2018026873 Feb 2018 WO
WO 2018045181 Mar 2018 WO
WO 2018064640 Apr 2018 WO
WO 2018085599 May 2018 WO
WO 2018089550 May 2018 WO
WO 2018091676 May 2018 WO
WO 2018136397 Jul 2018 WO
WO 2018136856 Jul 2018 WO
WO 2018144582 Aug 2018 WO
WO 2019165318 Aug 2018 WO
WO 2018175779 Sep 2018 WO
WO 2018209398 Nov 2018 WO
WO 2019023214 Jan 2019 WO
WO 2019032760 Feb 2019 WO
WO 2019068880 Apr 2019 WO
WO 2019113457 Jun 2019 WO
WO 2019126313 Jun 2019 WO
WO 2019140201 Jul 2019 WO
WO 2019213254 Nov 2019 WO
WO 2019213294 Nov 2019 WO
WO 2019241290 Dec 2019 WO
WO 2020028194 Feb 2020 WO
WO 2020047002 Mar 2020 WO
WO 2020047005 Mar 2020 WO
WO 2020047010 Mar 2020 WO
WO 2020053655 Mar 2020 WO
WO 2020056381 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 2020112604 Jun 2020 WO
WO 2020117914 Jun 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 2020167862 Aug 2020 WO
WO 2020176788 Sep 2020 WO
WO 2020176882 Sep 2020 WO
WO 2020190509 Sep 2020 WO
WO 2020206285 Oct 2020 WO
WO 2020219901 Oct 2020 WO
WO 2020240025 Dec 2020 WO
WO 2020243579 Dec 2020 WO
WO 2020254519 Dec 2020 WO
WO 2021041974 Mar 2021 WO
WO 2021067246 Apr 2021 WO
WO 2021091611 May 2021 WO
WO 2021092433 May 2021 WO
WO 2021097255 May 2021 WO
WO 2021116715 Jun 2021 WO
WO 2021119320 Jun 2021 WO
WO 2021133842 Jul 2021 WO
WO 2021133845 Jul 2021 WO
WO 2021133849 Jul 2021 WO
WO 2021168261 Aug 2021 WO
WO 2021168278 Aug 2021 WO
WO 2021207610 Oct 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 2021263111 Dec 2021 WO
WO 2022025965 Feb 2022 WO
WO 2022060798 Mar 2022 WO
WO 2022060953 Mar 2022 WO
WO 2022087273 Apr 2022 WO
WO 2022099037 May 2022 WO
WO 2022103712 May 2022 WO
WO 2022109181 May 2022 WO
WO 2022140028 Jun 2022 WO
WO 2022198068 Sep 2022 WO
WO 2022212269 Oct 2022 WO
WO 2022221425 Oct 2022 WO
WO 2022226057 Oct 2022 WO
WO 2022236054 Nov 2022 WO
WO 2022243303 Nov 2022 WO
WO 2022256503 Dec 2022 WO
WO 2022271820 Dec 2022 WO
WO 2023287765 Jan 2023 WO
WO 2023018799 Feb 2023 WO
WO 2023034489 Mar 2023 WO
WO 2023076345 May 2023 WO
WO 2023086880 May 2023 WO
WO 2023102118 Jun 2023 WO
WO 2023150098 Aug 2023 WO
WO 2023150163 Aug 2023 WO
WO 2023150171 Aug 2023 WO
WO 2023225519 Nov 2023 WO
Non-Patent Literature Citations (373)
Entry
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.
Dalma-Weiszhausz et al., “The Affymetrix GeneChip platform: an overview,” Methods Enzymol., 2006, 410:3-28.
Jensen et al., “Zinc fixation preserves flow cytometry scatter and fluorescence parameters and allows simultaneous analysis of DNA content and synthesis, and intracellular and surface epitopes,” Cytometry A., Aug. 2010, 77(8):798-804.
Lahiani et al., “Enabling Histopathological Annotations on Immunofluorescent Images through Virtualization of Hematoxylin and Eosin,” J Pathol Inform., Feb. 2018, 9:1, 8 pages.
Lou et al., “A review of room temperature storage of biospecimen tissue and nucleic acids for anatomic pathology laboratories and biorepositories,” Clin Biochem., Mar. 2014, 47(4-5):267-73.
Lykidis et al., “Novel zinc-based fixative for high quality DNA, RNA and protein analysis,” Nucleic Acids Res., Jun. 2007, 35(12):e85, 10 pages.
Miller et al., “Chapter 11—Solid and Suspension Microarrays for Microbial Diagnostics,” Methods in Microbiology, 2015, 42:395-431.
Nadji et al., “Immunohistochemistry of tissue prepared by a molecular-friendly fixation and processing system,” Appl Immunohistochem Mol Morphol., Sep. 2005, 13(3):277-82.
Passow et al., “RNAlater and flash freezing storage methods nonrandomly influence observed gene expression in RNAseq experiments,” bioRxiv, Jul. 2018, 28 pages.
Porreca et al., “Polony DNA sequencing,” Curr Protoc Mol Biol., Nov. 2006, Chapter 7, Unit 7.8, pp. 7.8.1-7.8.22.
Vickovic et al., “SM-Omics: An automated Platform for High-Throughput Spatial Multi-Omics,” bioRxiv, Oct. 2020, 40 pages.
Wohnhaas et al., “DMSO cryopreservation is the method of choice to preserve cells for droplet-based single-cell RNA sequencing,” Scientific Reports, Jul. 2019, 9(1):10699, 14 pages.
Fu et al., “Repeat subtraction-mediated sequence capture from a complex genome,” Plant J., Jun. 2010, 62(5):898-909.
Ganguli et al., “Pixelated spatial gene expression analysis from tissue,” Nat Commun., Jan. 2018, 9(1):202, 9 pages.
Hahnke et al., “Striptease on glass: validation of an improved stripping procedure for in situ microarrays,” J Biotechnol., Jan. 2007, 128(1):1-13.
Hu et al., “High reproducibility using sodium hydroxide-stripped long oligonucleotide DNA microarrays,” Biotechniques, Jan. 2005, 38(1):121-4.
Lee et al., “Improving the efficiency of genomic loci capture using oligonucleotide arrays for high throughput resequencing,” BMC Genomics, Dec. 2009, 10:646, 12 pages.
Schwers et al., “A high-sensitivity, medium-density, and target amplification-free planar waveguide microarray system for gene expression analysis of formalin-fixed and paraffin-embedded tissue,” Clin. Chem., Nov. 2009, 55(11):1995-2003.
Zahra et al., “Assessment of Different Permeabilization Methods of Minimizing Damage to the Adherent Cells for Detection of Intracellular RNA by Flow Cytometry,” Avicenna Journal of Medical Biotechnology, Jan. 1, 2014, 6(1):38-46.
U.S. Appl. No. 60/416,118, filed Oct. 3, 2002, Fan et al.
U.S. Appl. No. 61/267,363, filed Dec. 7, 2009, Fan et al.
[No Author Listed], “Chromium Next GEM Single Cell 3′ Reagent Kits v3.1,” User Guide, Document No. CG000204, 10x Genomics, Nov. 2019, 58 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).
[No Author Listed], “Proseek® Multiplex 96×96 User Manual,” Olink Proteomics, Olink Bioscience, Uppsala, Sweden, 2017, 20 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>, 70 pages.
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/5UJrN0cH17rEk0UXwdl9It/e54d99fb08a8f1500aba503005a04a56/CG000238_VisiumSpatialTissueOptimizationUserGuide_RevD.pdf>, 43 pages.
Adessi et al., “Solid phase DNA amplification: characterisation of primer attachment and amplification mechanisms,” Nucl. Acids Res., 2000, 28(20):E87, 8 pages.
Adiconis et al., “Comparative analysis of RNA sequencing methods for degraded or low-input samples,” Nat Methods, Jul. 2013, 10(7):623-9.
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.
Andresen et al., “Helicase-dependent amplification: use in OnChip amplification and potential for point-of-care diagnostics,” Expert Rev Mol Diagn., Oct. 2009, 9(7):645-650.
Archer et al., “Selective and flexible depletion of problematic sequences from RNA-seq libraries at the cDNA stage,” BMC Genomics, May 2014, 15(1):401, 9 pages.
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.
Bartosovic et al., “Single-cell CUT&Tag profiles histone modifications and transcription factors in complex tissues,” Nat Biotechnol., Jul. 2021, 39(7):825-835, Abstract.
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.
Bell, “A simple way to treat PCR products prior to sequencing using ExoSAP-IT,” Biotechniques, 2008, 44(6):834, 1 page.
Bentley et al., “Accurate whole human genome sequencing using reversible terminator chemistry,” Nature, 2008, 456(7218):53-59.
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.
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., “DNA hybridization detection in a microfluidic Channel using two fluorescently labelled nucleic acid probes,” Biosensors and Bioelectronics, 2008, 23(12):1878-1882.
Chen et al., “Expansion microscopy,” Science, 2015, 347(6221):543-548.
Chen et al., “Nanoscale imaging of RNA with expansion microscopy,” Nat Methods, Aug. 2016, 13(8):679-84.
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., “Spatially resolved, highly multiplexed RNA profiling in single cells,” Science, 2015, 348(6233):aaa6090, 21 pages.
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.
Chrisey et al., “Covalent attachment of synthetic DNA to self-assembled monolayer films,” Nucleic Acids Res., Aug. 1996, 24(15):3031-9.
Constantine et al., “Use of genechip high-density oligonucleotide arrays for gene expression monitoring,” Life Sceience News, Amersham Life Science, 1998, pp. 11-14.
Corces et al., “An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues,” Nat. Methods, 2017, 14(10):959-962.
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.
Cujec et al., “Selection of v-abl tyrosine kinase substrate sequences from randomized peptide and cellular proteomic libraries using mRNA display,” Chemistry and Biology, 2002, 9(2):253-264.
Dahl et al., “Circle-to-circle amplification for precise and sensitive DNA analysis,” Proc. Natl. Acad. Sci., 2004, 101(13):4548-4553.
Darmanis et al., “ProteinSeq: High-Performance Proteomic Analyses by Proximity, Ligation and Next Generation Sequencing,” PLos One, 2011, 6(9):e25583, 10 pages.
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.
Deamer et al., “Characterization of nucleic acids by nanopore analysis,” Acc Chem Res., Oct. 2002, 35(10):817-25.
Dean et al., “Comprehensive human genome amplification using multiple displacement amplification,” Proc Natl. Acad. Sci. USA, 2002, 99(8):5261-66.
Dressman et al., “Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations,” Proc. Natl. Acad. Sci. USA, 2003, 100(15):8817-8822.
Drmanac et al., “CoolMPS™: Advanced massively parallel sequencing using antibodies specific to each natural nucleobase,” BioRxiv, 2020, 19 pages.
Druley et al., “Quantification of rare allelic variants from pooled genomic DNA,” Nat. Methods, 2009, 6(4):263-65.
Duncan et al., “Affinity chromatography of a sequence-specific DNA binding protein using Teflon-linked oligonucleotides,” Anal. Biochem., 1988, 169(1):104-108.
Eberwine, “Amplification of mRNA populations using aRNA generated from immobilized oligo(dT)-T7 primed cDNA,” BioTechniques, 1996, 20(4):584-91.
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.
Eng et al., “Profiling the transcriptome with RNA SPOTs,” Nat Methods., 2017, 14(12):1153-1155.
Fire et al., “Rolling replication of short DNA circles,” Proc. Natl. Acad. Sci., 1995, 92(10):4641-4645.
Flanigon et al., “Multiplex protein detection with DNA readout via mass spectrometry,” N. Biotechnol., 2013, 30(2):153-158.
Fluidigm, “Equivalence of Imaging Mass Cytometry and Immunofluorescence on FFPE Tissue Sections,” White Paper, 2017, 12 pages.
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.
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.
Gill et al., “Nucleic acid isothermal amplification technologies: a review,” Nucleosides Nucleotides Nucleic Acids, Mar. 2008, 27(3):224-43.
Glass et al., “SIMPLE: a sequential immunoperoxidase labeling and erasing method,” J. Histochem. Cytochem., Oct. 2009, 57(10):899-905.
Gloor, “Gene targeting in Drosophila,” Methods Mol Biol., 2004, 260:97-114.
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.
Goryshin et al., “Tn5 in vitro transposition,” J Biol Chem., Mar. 1998, 273(13):7367-74.
Gracia Villacampa et al., “Genome-wide Spatial Expression Profiling in FFPE Tissues,” bioRxiv, 2020, pp. 38 pages.
Gu et al., “Protein tag-mediated conjugation of oligonucleotides to recombinant affinity binders for proximity ligation,” N Biotechnol., 2013, 30(2):144-152.
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.
Hardenbol et al., “Highly multiplexed molecular inversion probe genotyping: over 10,000 targeted SNPs genotyped in a single tube assay,” Genome Res., Feb. 2005, 15(2):269-75.
Hardenbol et al., “Multiplexed genotyping with sequence-tagged molecular inversion probes,” Nature Biotechnol., Jun. 2003, 21(6):673-678.
He et al., “In situ synthesis of protein arrays,” Current Opinion in Biotechnology, 2008, 19(1):4-9.
He et al., “Printing protein arrays from DNA arrays,” Nature Methods, 2008, 5(2):175-77.
He, “Cell-free protein synthesis: applications in proteomics and biotechnology,” New Biotechnology, 2008, 25(2-3):126-132.
Healy, “Nanopore-based single-molecule DNA analysis,” Nanomedicine (Lond), Aug. 2007, 2(4):459-81.
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.
Ho et al., “Bacteriophage T4 RNA ligase 2 (gp24.1) exemplifies a family of RNA ligases found in all phylogenetic domains,” PNAS, Oct. 2002, 99(20):12709-14.
Hycultbiotech.com, [online], “Immunohistochemistry, Paraffin” Apr. 2010, retrieved on Apr. 16, 2020, retrieved from URL<https://www.hycultbiotech.com/media/wysiwyg/Protocol_Immunohistochemistry_Paraffin_2.pdf>, 3 pages.
Ichikawa et al., “In vitro transposition of transposon Tn3,” J Biol. Chem., Nov. 1990, 265(31):18829-32, Abstract.
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.
Kap et al., “Histological assessment of PAXgene tissue fixation and stabilization reagents,” PLoS One, 2011, 6:e27704, 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.
Karmakar et al., “Organocatalytic removal of formaldehyde adducts from RNA and DNA bases,” Nature Chemistry, Aug. 3, 2015, 7(9):752-758.
Kaya-Okur et al., “CUT&Tag for efficient epigenomic profiling of small samples and single cells,” Apr. 2019, 10(1):1930, 10 pages.
Kennedy-Darling et al., “Measuring the Formaldehyde Protein-DNA Cross-Link Reversal Rate,” Analytical Chemistry, 2014, 86(12):5678-5681.
Kent et al., “Polymerase θ is a robust terminal transferase that oscillates between three different mechanisms during end-joining” Elife, Jun. 2016, 5:e13740, 25 pages.
Kirby et al., “Cryptic plasmids of Mycobacterium avium: Tn552 to the rescue,” Mol Microbiol., Jan. 2002, 43(1):173-86.
Kleckner et al., “Tn10 and IS10 transposition and chromosome rearrangements: mechanism and regulation in vivo and in vitro,” Curr Top Microbiol Immunol., 1996, 204:49-82.
Korbel et al., “Paired-end mapping reveals extensive structural variation in the human genome,” Science, 2007, 318(5849):420-426.
Kozlov et al., “A highly scalable peptide-based assay system for proteomics,” PLoS One, 2012, 7(6):e37441, 10 pages.
Kozlov et al., “A method for rapid protease substrate evaluation and optimization,” Comb Chem High Throughput Screen, 2006, 9(6):481-87.
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.
Lampe et al., “A purified mariner transposase is sufficient to mediate transposition in vitro,” Embo J., Oct. 1996, 15(19):5470-9.
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.
Larman et al., “Sensitive, multiplex and direct quantification of RNA sequences using a modified RASL assay,” Nucleic Acids Research, 2014, 42(14):9146-9157.
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.
Li et al., “A photocleavable fluorescent nucleotide for DNA sequencing and analysis,” Proc. Natl. Acad. Sci., 2003, 100(2):414-419.
Li et al., “RASL-seq for Massively Parallel and Quantitative Analysis of Gene Expression,” Curr Protoc Mol Biol., Apr. 2012, 4(13):1-10.
Li et al., “Review: a comprehensive summary of a decade development of the recombinase polymerase amplification,” Analyst, Dec. 2018, 144(1):31-67.
Lin et al., “Highly multiplexed imaging of single cells using a high-throughput cyclic immunofluorescence method,” Nat Commun., Sep. 2015, 6:8390, 7 pages.
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.
Lovatt et al., “Transcriptome in vivo analysis (TIVA) of spatially defined single cells in live tissue,” Nature Methods, 2013, 11(2):190-196.
Lund et al., “Assessment of methods for covalent binding of nucleic acids to magnetic beads, Dynabeads, and the characteristics of the bound nucleic acids in hybridization reactions,” Nucleic Acids Res., 1988, 16(22):10861-80.
Lundberg et al., “High-fidelity amplification using a thermostable DNA polymerase isolated from Pyrococcus furiosus,” Gene, 1991, 108(1):1-6.
Lundberg et al., “Homogeneous antibody-based proximity extension assays provide sensitive and specific detection of low-abundant proteins in human blood,” Nucleic Acids Res., 2011, 39(15):e102, 8 pages.
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.
Lundin et al., “Increased throughput by parallelization of library preparation for massive sequencing,” PLoS One, Apr. 2010, 5(4):e10029, 7 pages.
Lyck et al., “Immunohistochemical markers for quantitative studies of neurons and glia in human neocortex,” J Histochem Cytochem, 2008, 56(3):201-21.
MacIntyre, “Unmasking antigens for immunohistochemistry.,” Br J Biomed Sci., 2001, 58(3):190-6.
Marx, “Method of the Year: spatially resolved transcriptomics,” Nature Methods, 2021, 18(1):9-14.
McCloskey et al., “Encoding PCR products with batch-stamps and barcodes,” Biochem. Genet., 2007, 45(11-12):761-767.
Meers et al., “Improved CUT&RUN chromatin profiling tools,” Elife, Jun. 2019, 8:e46314, 16 pages.
Merritt et al., “Multiplex digital spatial profilin gof 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.
Miele et al., “Mapping cis- and trans- chromatin interaction networks using chromosome conformation capture (3C),” Methods Mol Biol., 2009, 464:105-21.
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.
Motea et al., “Terminal deoxynucleotidyl transferase: the story of a misguided DNA polymerase,” Biochim Biophys Acta., May 2010, 1804(5):1151-66.
Nandakumar et al., “How an RNA Ligase Discriminates RNA versus DNA Damage,” Molecular Cell, 2004, 16:211-221.
Ncbi.nlm.nih.gov, [online], “Molecular Inversion Probe Assay,” available on or before Oct. 14, 2014, via Internet Archive: Wayback Machine URL<https://web.archive.org/web/20141014124037/https://www.ncbi.nlm.nih.gov/probe/docs/techmip/>, retrieved on Jun. 16, 2021, retrieved from URL<https://www.ncbi.nlm.nih.gov/probe/docs/techmip/>, 2 pages.
Ng et al., “Gene identification signature (GIS) analysis for transcriptome characterization and genome annotation,” Nature Methods, 2005, 2(2):105-111.
Nichols et al., “RNA Ligases,” Curr Protoc Mol Biol., Oct. 2008, 84(1):3.15.1-3.15.4.
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.
Niklas et al., “Selective permeabilization for the high-throughput measurement of compartmented enzyme activities in mammalian cells,” Anal Biochem, Sep. 2011, 416(2):218-27.
Nowak et al., “Entering the Postgenome Era,” Science, 1995, 270(5235):368-71.
Ohtsubo et al., “Bacterial insertion sequences,” Curr Top Microbiol Immunol., 1996, 204:1-26.
Pandey et al., “Inhibition of terminal deoxynucleotidyl transferase by adenine dinucleotides. Unique inhibitory action of Ap5A, ” FEBS Lett., Mar. 1987, 213(1):204-8.
Park et al., “Single cell trapping in larger microwells capable of supporting cell spreading and proliferation,” Microfluid Nanofluid, 2010, 8:263-268.
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.
Picelli et al., “Tn5 transposase and tagmentation procedures for massively scaled sequencing projects,” Genome Res., Dec. 2014, 24(12):2033-40.
Pipenburg et al., “DNA detection using recombination proteins,” PLOS Biol., Jul. 2006, 4(7):e204, 7 pages.
Plasterk, “The Tc1/mariner transposon family,” Curr Top Microbiol Immunol., 1996, 204:125-43.
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.
U.S. Appl. No. 61/267,363, filed Dec. 7, 2009, 33 pages.
Raab et al., “Human tRNA genes function as chromatin insulators,” EMBO J., Jan. 2012, 31(2):330-50.
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.
Reznikoff, “Tn5 as a model for understanding DNA transposition,” Mol Microbiol., Mar. 2003, 47(5):1199-206.
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.
Roy et al., “Assessing long-distance RNA sequence connectivity via RNA-templated DNA-DNA ligation,” eLife, 2015, 4:e03700, 21 pages.
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.
Shelbourne et al., “Fast copper-free click DNA ligation by the ring-strain promoted alkyne-azide cycloaddition reaction,” Chem. Commun., 2011, 47(22):6257-6259.
Shendure et al., “Accurate multiplex polony sequencing of an evolved bacterial genome,” Science, 2005, 309(5741):1728-1732.
Simonis et al., “Nuclear organization of active and inactive chromatin domains uncovered by chromosome conformation capture-on-chip (4C),” Nat Genet., Nov. 2006, 38(11):1348-54.
Skene et al., “An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites,” Elife, Jan. 2017, 6:e21856, 35 pages.
Spitale et al., “Structural imprints in vivo decode RNA regulatory mechanisms,” Nature, 2015, 519(7544):486-90.
Stahl et al., “Visualization and analysis of gene expression in tissue sections by spatial transcriptomics,” Science, Jun. 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.
Stoddart et al., “Single-nucleotide discrimination in immobilized DNA oligonucleotides with a biological nanopore,” PNAS USA., May 2009, 106(19):7702-7707.
Strell et al., “Placing RNA in context and space—methods for spatially resolved transcriptomics,” The FEBS Journal, 2019, 286(8):1468-1481.
Stroh et al., “Quantum dots spectrally distinguish multiple species within the tumor milieu in vivo,” Nat Med., Jun. 2005, 11(6):678-82.
Taylor et al., “Mitochondrial DNA mutations in human disease,” Nature Reviews Genetics, May 2005, 6(5):389-402.
Tentori et al., “Detection of Isoforms Differing by a Single Charge Unit in Individual Cells,” Chem. Int. Ed., 2016, 55(40):12431-5.
Tian et al., “Antigen peptide-based immunosensors for rapid detection of antibodies and antigens,” Anal Chem, 2009, 81(13):5218-5225.
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.
Tolbert et al., “New methods for proteomic research: preparation of proteins with N-terminal cysteines for labeling and conjugation,” Angewandte Chemie International Edition, Jun. 2002, 41(12):2171-4.
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.
U.S. Appl. No. 60/416,118 Fan et al., Multiplex Nucleic Acid Analysis Using Archived or Fixed Samples, filed Oct. 3, 2002, 22 pages.
Van Gelder et al., “Amplified RNA synthesized from limited quantities of heterogeneous cDNA,” Proc. Natl. Acad. Sci. USA, 1990, 87(5):1663-1667.
Vandernoot et al., “cDNA normalization by hydroxyapatite chromatography to enrich transcriptome diversity in RNA-seq applications,” Biotechniques, Dec. 2012, 53(6):373-80.
Vasiliskov et al., “Fabrication of microarray of gel-immobilized compounds on a chip by copolymerization,” Biotechniques, Sep. 1999, 27(3):592-606.
Velculescu et al., “Serial analysis of gene expression,” Science, Oct. 1995, 270(5235):484-7.
Vickovic et al., “High-definition spatial transcriptomics for in situ tissue profiling,” Nature Methods, 2019, 9 pages.
Vincent et al., “Helicase-dependent isothermal DNA amplification,” EMBO Rep., Aug. 2004, 5(8):795-800.
Viollet et al., “T4 RNA ligase 2 truncated active site mutants: improved tools for RNA analysis, ” BMC Biotechnol., Jul. 2011, 11:72, 14 pages.
Vogelstein et al., “Digital PCR,” Proceedings of the National Academy of Sciences, Aug. 1999, 96(16):9236-9241.
Waichman et al., “Functional immobilization and patterning of proteins by an enzymatic transfer reaction,” Analytical chemistry, 2010, 82(4):1478-85.
Walker et al., “Strand displacement amplification—an isothermal, in vitro DNA amplification technique,” Nucleic Acids Research, 1992, 20(7):1691-1696.
Wang et al., “Concentration gradient generation methods based on microfluidic systems, ” RSC Adv., 2017, 7:29966-29984.
Wang et al., “High-fidelity mRNA amplification for gene profiling,” Nature Biotechnology, Apr. 2000, 18(4):457-459.
Wang, “RNA amplification for successful gene profiling analysis,” J Transl Med., Jul. 2005, 3:28, 11 pages.
Weinreich et al., “Evidence that the cis Preference of the Tn5 Transposase is Caused by Nonproductive Multimerization,” Genes and Development, Oct. 1994, 8(19):2363-2374.
Wilson et al., “New transposon delivery plasmids for insertional mutagenesis in Bacillus anthracis,” J Microbiol Methods, Dec. 2007, 71(3):332-5.
Wolf et al., “Rapid hybridization kinetics of DNA attached to submicron latex particles,” Nucleic Acids Res, 1987, 15(7):2911-2926.
Wong et al., “Direct Site-Selective Covalent Protein Immobilization Catalyzed by a Phosphopantetheinyl Transferase,” J. Am. Chem Soc., 2008, 130(37):12456-64.
Worthington et al., “Cloning of random oligonucleotides to create single-insert plasmid libraries,” Anal Biochem, 2001, 294(2):169-175.
Wu et al., “Detection DNA Point Mutation with Rolling-Circle Amplification Chip,” IEEE, 2010 4th International Conference on Bioinformatics and Biomedical Engineering, Jun. 2010, 4 pages.
Yeakley et al, “Profiling alternative splicing on fiber-optic arrays,” Nature biotechnology, 2002, 20:353-358.
Yeakley et al., “A trichostatin A expression signature identified by TempO-Seq targeted whole transcriptome profiling,” PLoS One, May 2017, 12(5):e0178302, 22 pages.
Yershov et al., “DNA analysis and diagnostics on oligonucleotide microchips,” Proc. Natl. Acad. Sci. USA, May 1996, 93(10):4913-4918.
Yin et al., “Genetically encoded short peptide tag for versatile protein labeling by Sfp phosphopantetheinyl transferase,” PNAS, 2005, 102(44):15815-20.
Zhang et al., “Archaeal RNA ligase from thermoccocus kodakarensis for template dependent ligation,” RNA Biol., Jan. 2017, 14(1):36-44.
Zhang et al., “Assembling DNA through Affinity Binding to Achieve Ultrasensitive Protein Detection,” Angew Chem Int Ed Engl., 2013, 52(41):10698-705.
Zhang et al., “Binding-induced DNA assembly and its application to yoctomole detection of proteins,” Anal Chem, 2012, 84(2):877-884.
Zhang et al., “Multiplex ligation-dependent probe amplification (MLPA) for ultrasensitive multiplexed microRNA detection using ribonucleotide-modified DNA probes†,” Chem. Commun., 2013, 49:10013-10015.
Zheng et al., “Origins of human mitochondrial point mutations as DNA polymerase gamma-mediated errors,” Mutat. Res., 2006, 599(1-2):11-20.
Zhou et al., “Genetically encoded short peptide tags for orthogonal protein labeling by Sfp and AcpS phosphopantetheinyl transferases,” ACS Chemical Biol., 2007, 2(5):337-346.
Zhu et al., “Reverse transcriptase template switching: a Smart approach for full-length cDNA library construction,” Biotechniques, Apr. 2001, 30(4):892-897.
Aran et al., “xCell: digitally portraying the tissue cellular heterogeneity landscape,” Genome Biol., Nov. 2017, 18(1):220, 14 pages.
Plongthongkum et al., “Advances in the profiling of DNA modifications: cytosine methylation and beyond,” Nature Reviews Genetics, Aug. 2014, 15(10):647-661.
Salmén et al., “Barcoded solid-phase RNA capture for Spatial Transcriptomics profiling in mammalian tissue sections,” Nature Protocols, Oct. 2018, 13(11):2501-2534.
Hayes et al., “Electrophoresis of proteins and nucleic acids: I-Theory,” BMJ, Sep. 1989, 299(6703):843-6.
Grokhovsky, “Specificity of DNA cleavage by ultrasound,” Molecular Biology, 2006, 40(2):276-283.
Schweitzer et al., “Multiplexed protein profiling on microarrays by rolling-circle amplification,” Nature Biotechnology, Apr. 2002, 20(4):359-365.
Asp et al., “Spatially Resolved Transcriptomes-Next Generation Tools for Tissue Exploration,” Bioessays, Oct. 2020, 42(10):e1900221, 16 pages.
Balakrishnan et al., “Flap endonuclease 1,” Annu Rev Biochem., Jun. 2013, 82:119-138.
Bergenstråhle et al., “Seamless integration of image and molecular analysis for spatial transcriptomics workflows,” BMC Genomics, Jul. 2020, 21(1):482, 7 pages.
Berger et al., “Universal bases for hybridization, replication and chain termination,” Nucleic Acid Res., Aug. 2000, 28(15):2911-2914.
Blair et al., “Microarray temperature optimization using hybridization kinetics,” Methods Mol Biol., 2009, 529:171-96.
Bolotin et al., “MiXCR: software for comprehensive adaptive immunity profiling, ” Nat Methods., May 2015, 12(5):380-1.
Chen et al., “Parallel single nucleotide polymorphism genotyping by surface invasive cleavage with universal detection,” Anal Chem., Apr. 2005, 77(8):2400-5.
Chester et al., “Dimethyl sulfoxide-mediated primer Tm reduction: a method for analyzing the role of renaturation temperature in the polymerase chain reaction,” Anal Biochem, Mar. 1993, 209(2):284-90.
Ciaccio et al., “Systems analysis of EGF receptor signaling dynamics with microwestern arrays,” Nat Methods, Feb. 2010, 7(2):148-55.
Gerard et al., “Excess dNTPs minimize RNA hydrolysis during reverse transcription,” Biotechniques, Nov. 2002, 33(5):984, 986, 988, 990.
Hessner et al., “Genotyping of factor V G1691A (Leiden) without the use of PCR by invasive cleavage of oligonucleotide probes,” Clin Chem., Aug. 2000, 46(8 Pt 1):1051-6.
Hoffman et al., “Formaldehyde crosslinking: a tool for the study of chromatin complexes,” J Biol Chem., Oct. 2015, 290(44):26404-11.
Hughes et al., “Microfluidic Western blotting,” PNAS, Dec. 2012, 109(52):21450-21455.
Jucá et al., “Effect of dimethyl sulfoxide on reverse transcriptase activity,” Braz. J. Med. Biol. Res., Mar. 1995, 28(3):285-90.
Kalantari et al., “Deparaffinization of formalin-fixed paraffin-embedded tissue blocks using hot water instead of xylene,” Anal Biochem., Aug. 2016, 507:71-3.
Krzywkowski et al., “Chimeric padlock and iLock probes for increased efficiency of targeted RNA detection,” RNA, Jan. 2019, 25(1):82-89.
Krzywkowski et al., “Fidelity of RNA templated end-joining by chlorella virus DNA ligase and a novel iLock assay with improved direct RNA detection accuracy,” Nucleic Acids Research, Oct. 2017, 45(18):e161, 9 pages.
Lyamichev et al., “Invader assay for SNP genotyping,” Methods Mol Biol., 2003, 212:229-40.
Lyamichev et al., “Polymorphism identification and quantitative detection of genomic DNA by invasive cleavage of oligonucleotide probes,” Nat Biotechnol., Mar. 1999, 17(3):292-6.
Nilsson et al., “RNA-templated DNA ligation for transcript analysis,” Nucleic Acids Res., Jan. 2001, 29(2):578-81.
Olivier, “The Invader assay for SNP genotyping,” Mutat. Res., Jun. 2005, 573(1-2):103-110.
Penno et al., “Stimulation of reverse transcriptase generated cDNAs with specific indels by template RNA structure: retrotransposon, dNTP balance, RT-reagent usage,” Nucleic Acids Res., Sep. 2017, 45(17):10143-10155.
Perocchi et al., “Antisense artifacts in transcriptome microarray experiments are resolved by actinomycin D,” Nucleic Acids Res., 2007, 35(19):e128, 7 pages.
Picelli et al., “Full-length RNA-seq from single cells using Smart-seq2,” Nat Protoc., Jan. 2014, 9(1):171-81.
Schouten et al., “Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification,” Nucleic Acids Res., Jun. 2002, 30(12):e57, 13 pages.
Singh et al., “High-throughput targeted long-read single cell sequencing reveals the clonal and transcriptional landscape of lymphocytes,” Nat Commun., Jul. 2019, 10(1):3120, 13 pages.
Sountoulidis et al., “SCRINSHOT, a spatial method for single-cell resolution mapping of cell states in tissue sections,” PLoS Biol., Nov. 2020, 18(11):e3000675, 32 pages.
Spiess et al., “A highly efficient method for long-chain cDNA synthesis using trehalose and betaine,” Anal. Biochem., Feb. 2002, 301(2):168-74.
Sutherland et al., “Utility of formaldehyde cross-linking and mass spectrometry in the study of protein-protein interactions,” J. Mass Spectrom., Jun. 2008, 43(6):699-715.
Toubanaki et al., “Dry-reagent disposable biosensor for visual genotyping of single nucleotide polymorphisms by oligonucleotide ligation reaction: application to pharmacogenetic analysis,” Hum Mutat., Aug. 2008, 29(8):1071-8.
Tu et al., “TCR sequencing paired with massively parallel 3′ RNA-seq reveals clonotypic T cell signatures,” Nature Immunology, Dec. 2019, 20(12):1692-1699.
Vázquez Bernat et al., “High-Quality Library Preparation for NGS-Based Immunoglobulin Germline Gene Inference and Repertoire Expression Analysis,” Front Immunol., Apr. 2019, 10:660, 12 pages.
Wu et al., “RollFISH achieves robust quantification of single-molecule RNA biomarkers in paraffin-embedded tumor tissue samples, ” Commun Biol., Nov. 2018, 1:209, 8 pages.
Yasukawa et al., “Effects of organic solvents on the reverse transcription reaction catalyzed by reverse transcriptases from avian myeloblastosis virus and Moloney murine leukemia virus,” Biosci Biotechnol Biochem., 2010, 74(9):1925-30.
Bibikova et al., “Quantitative gene expression profiling in formalin-fixed paraffin-embedded tissues using universal bead arrays,” The American Journal of Pathology, Nov. 1, 2004, 165(5):1799-1807.
Choi et al., “Multiplexed detection of mRNA using porosity-tuned hydrogel microparticles,” Analytical chemistry, Sep. 28, 2012, 84(21):9370-9378.
Fan et al., “A versatile assay for high-throughput gene expression profiling on universal array matrices, ” Genome Research, May 1, 2004, 14(5):878-885.
Alam, “Proximity Ligation Assay (PLA),” Curr Protoc Immunol., Nov. 2018, 123(1):e58, 8 pages.
Li et al., “An activity-dependent proximity ligation platform for spatially resolved quantification of active enzymes in single cells,” Nat Commun, Nov. 2017, 8(1):1775, 12 pages.
Ristic et al., “Detection of Protein-Protein Interactions and Posttranslational Modifications Using the Proximity Ligation Assay: Application to the Study of the SUMO Pathway, ” Methods Mol. Biol., 2016, 1449:279-90.
Wang et al., “Imaging-based pooled CRISPR screening reveals regulators of IncRNA localization,” Proc Natl Acad Sci USA, May 2019, 116(22):10842-10851.
Wiedmann et al., “Ligase chain reaction (LCR)—overview and applications,” PCR Methods Appl., Feb. 1994, 3(4):S51-64.
Appella, “Non-natural nucleic acids for synthetic biology,” Current Opinion in Chemical Biology, Dec. 2009, 13(5-6): 687-696.
Bunt et al., “FRET from single to multiplexed signaling events,” Biophys Rev. Apr. 2017, 9(2): 119-129.
Grünweller et al., “Locked Nucleic Acid Oligonucleotides,” BioDrugs, Jul. 2007, 21(4): 235-243.
Gu et al., “Multiplex single-molecule interaction profiling of DNA-barcoded proteins,” Nature, Sep. 21, 2014, 515:554-557.
Ma et al., “Isothermal amplification method for next-generation sequencing,” PNAS, Aug. 12, 2013, 110(35):14320-14323.
Orenstein et al., “γPNA FRET Pair Miniprobes for Quantitative Fluorescent In Situ Hybridization to Telomeric DNA in Cells and Tissue,” Molecules, Dec. 2, 2017, 22(12):2117, 15 pages.
Illumina.com [online], “Ribo-Zero® rRNA Removal Kit Reference Guide,” Aug. 2016, retrieved on Apr. 26, 2022, retrieved from URL<https://jp.support.illumina.com/content/dam/illumina-support/documents/documentation/chemistry_documentation/ribosomal-depletion/ribo-zero/ribo-zero-reference-guide-15066012-02.pdf>, 36 pages.
Niedringhaus et al., “Landscape of next-generation sequencing technologies,” Anal Chem., Jun. 2011, 83(12):4327-41.
Qiu et al., “Combination probes with intercalating anchors and proximal fluorophores for DNA and RNA detection,” Nucleic Acids Research, Sep. 2016, 44(17):e138, 12 pages.
Zhao et al., “Isothermal Amplification of Nucleic Acids,” Chemical Reviews, Nov. 2015, 115(22):12491-12545.
Goldmeyer et al., “Development of a novel one-tube isothermal reverse transcription thermophilic helicase-dependent amplification platform for rapid RNA detection,” Journal of Molecular Diagnostics, American Society for Investigative Pathology and the Association for Molecular Pathology, Nov. 1, 2007, 9(5):639-644.
[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.
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/SUJrN0cH17rEk0UXwdl9It/e54d99fb08a8f1500aba503005a04a56/CG000238_VisiumSpatialTissueOptimizationUserGuide_RevD.pdf>, 43 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/3GGIfH3RWpd1bFVha1pexR/8baa08d9007157592b65b2cdc7130990/CG000239_VisiumSpatialGeneExpression_UserGuide_RevD.pdf>, 70 pages.
Blanco et al., “A practical approach to FRET-based PNA fluorescence in situ hybridization,” Methods, Dec. 2010, 52(4):343-51.
Czarnik, “Encoding methods for combinatorial chemistry,” Curr Opin Chem Biol., Jun. 1997, 1(1):60-6.
MacBeath et al., “Printing proteins as microarrays for high-throughput function determination,” Science, Sep. 2000, 289(5485):1760-1763.
Morlan et al., “Selective depletion of rRNA enables whole transcriptome profiling of archival fixed tissue,” PLoS One, Aug. 2012, 7(8):e42882, 8 pages.
Pellestor et al., “The peptide nucleic acids (PNAs), powerful tools for molecular genetics and cytogenetics,” Eur J Hum Genet., Sep. 2004, 12(9):694-700.
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/4q03w6959AJFxffSw5lee9/6a2ac61cf6388a72564eeb96bc294967/CG000238_VisiumSpatialTissueOptimizationUserGuide_Rev_A.pdf>, 46 pages.
Barnett et al., “ATAC-Me Captures Prolonged DNA Methylation of Dynamic Chromatin Accessibility Loci during Cell Fate Transitions,” Mol Cell., Mar. 2020, 77(6):1350-1364.e6.
Cruz et al., “Methylation in cell-free DNA for early cancer detection,” Ann Oncol., Jun. 2018, 29(6):1351-1353.
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.
Ke et al., “In situ sequencing for RNA analysis in preserved tissue and cells,” Nat Methods., Sep. 2013, Supplementary Materials, 29 pages.
Miner et al., “Molecular barcodes detect redundancy and contamination in hairpin-bisulfite PCR,” Nucleic Acids Res., Sep. 2004, 32(17):e135, 4 pages.
Wang et al., “Tagmentation-based whole-genome bisulfite sequencing, ” Nature Protocols, Oct. 2013, 8(10):2022-2032.
U.S. Appl. No. 63/033,348, filed Jun. 2, 2020, Bent.
Arslan et al., “Engineering of a superhelicase through conformational control (Supplementary Materials),” Science, Apr. 17, 2015, 348(6232):344-347, 18 pages.
Arslan et al., “Engineering of a superhelicase through conformational control,” Science, Apr. 17, 2015, 348(6232):344-347.
Baner et al., “Signal amplification of padlock probes by rolling circle replication,” Nucleic Acids Res., 1998, 26(22):5073-5078.
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., “Efficient in situ barcode sequencing using padlock probe-based BaristaSeq,” Nucleic Acids Res., 2018, 46(4): e22, 11 pages.
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.
Dean et al., “Rapid Amplification of Plasmid and Phage DNA Using Phi29 DNA Polymerase and Multiply-Primed Rolling Circle Amplification,” Genome Research, Jun. 2001, 11:1095-1099.
Eng et al., “Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH+,” Nature, Apr. 2019, 568(7751):235-239, 37 pages.
Ergin et al., “Proteomic Analysis of PAXgene-Fixed Tissues,” J Proteome Res., 2010, 9(10):5188-96.
Faruqi et al., “High-throughput genotyping of single nucleotide polymorphisms with rolling circle amplification,” BMC Genomics, Aug. 2001, 2:4, 10 pages.
Gao et al., “A highly homogeneous expansion microscopy polymer composed of tetrahedron-like monomers,” bioRxiv, Oct. 22, 2019, 23 pages (Preprint).
Gilar et al., “Study of phosphorothioate-modified oligonucleotide resistance to 3′-exonuclease using capillary electrophoresis,” J Chromatogr B Biomed Sci Appl., Aug. 28, 1998, 714(1):13-20.
Goh et al., “Highly Specific Multiplexed RNA Imaging in Tissues With Split-FISH,” Nat Methods, Jun. 15, 2020, 17(7):689-693, 21 pages.
Goransson et al., “A single molecule array for digital targeted molecular analyses,” Nucleic Acids Res., Nov. 25, 2009, 37(1):e7, 9 pages.
Li et al., “A new GSH-responsive prodrug of 5-aminolevulinic acid for photodiagnosis and photodynamic therapy of tumors,” European Journal of Medicinal Chemistry, Nov. 2019, 181:111583, 9 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.
Mathieson et al., “A Critical Evaluation of the PAXgene Tissue Fixation System: Morphology, Immunohistochemistry, Molecular Biology, and Proteomics,” Am J Clin Pathol., Jul. 8, 2016, 146(1):25-40.
Mignardi et al., “Oligonucleotide gap-fill ligation for mutation detection and sequencing in situ,” Nucleic Acids Research, Aug. 3, 2015, 43(22):e151, 12 pages.
Mohsen et al., “The Discovery of Rolling Circle Amplification and Rolling Circle Transcription,” Acc Chem Res., Nov. 15, 2016, 49(11):2540-2550, 25 pages.
Nallur et al., “Signal amplification by rolling circle amplification on DNA microarrays,” Nucleic Acids Res., Dec. 1, 2001, 29(23):e118, 9 pages.
Raj et al., “Imaging individual mRNA molecules using multiple singly labeled probes,” Nature Methods, Oct. 2008, 5(10):877-879, 9 pages.
Schweitzer et al., “Immunoassays with rolling circle DNA amplification: A versatile platform for ultrasensitive antigen detection,” Proc. Natl Acad. Sci. USA, May 22, 2000, 97:10113-119.
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.
Howell et al., “iFRET: An Improved Fluorescence System for DNA-Melting Analysis,” Genome Research, 2002, 12:1401-1407.
Nam et al., “Nanoparticle-Based Bio-Bar Codes for the Ultrasensitive Detection of Proteins,” Science, Sep. 26, 2003, 301(5641):1884-1886.
Redmond et al., “Single-cell TCRseq: paired recovery of entire T-cell alpha and beta chain transcripts in T-cell receptors from single-cell RNAseq,” Genome Med, 2016, 8:80, 12 pages.
Belaghzal et al., “Hi-C 2.0: An Optimized Hi-C Procedure for High-Resolution Genome-Wide Mapping of Chromosome Conformation,” Methods, Jul. 1, 2017, 123:56-65, 20 pages.
Belton et al., “Hi-C: A comprehensive technique to capture the conformation of genomes,” Methods, Nov. 2012, 58(3):268-276, 16 pages.
Bentzen et al., “Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes,” Nat Biotechnol., Oct. 2016, 34(10):1037-1045, 12 pages.
Chen et al. “Arrayed profiling of multiple glycans on whole living cell surfaces.” Analytical chemistry, Oct. 15, 2013, 85(22):11153-11158.
Fan et al., “Illumina Universal Bead Arrays,” Methods in Enzymology, 2006, 410:57-73.
Hadrup et al., “Parallel detection of antigen-specific T-cell responses by multidimensional encoding of MHC multimers,” Nat. Methods., Jul. 2009, 6(7), 520-526.
Hobro et al, “An evaluation of fixation methods: Spatial and compositional cellular changes observed by Raman imaging,” Vibrational Spectroscopy, Jul. 2017, 91:31-45.
Landegren et al., “A Ligase-Mediated Gene Detection Technique,” Science, 1988, 241(4869):1077-1080.
Mamedov et al., “Preparing unbiased T-cell receptor and antibody cDNA libraries for the deep next generation sequencing profiling,” Frontiers in Immunol., Dec. 23, 2013, 4(456):1-10.
Oksuz et al., “Systematic evaluation of chromosome conformation capture assays,” Nature Methods, Sep. 2021, 18:1046-1055.
Rohland et al., “Partial uracil-DNA-glycosylase treatment for screening of ancient DNA,” Phil. Trans. R. Soc. B, Jan. 19, 2015, 370(1660): Jun. 24, 2013, 11 pages.
Schmidl et al., “ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors,” Nature Methods, Oct. 2015, 12:963-965.
Su et al., “Restriction enzyme selection dictates detection range sensitivity in chromatin conformation capture-based variant-to-gene mapping approaches,” bioRxiv, Dec. 15, 2020, 22 pages.
Sun et al., “Statistical Analysis of Spatial Expression Pattern for Spatially Resolved Transcriptomic Studies,” Nature Methods, Jan. 27, 2020, 17(2): 193-200.
Svensson et al., “SpatialDE: identification of spatially variable genes,” Nature Methods, May 2018, 15:343-346, 15 pages.
Related Publications (1)
Number Date Country
20210262019 A1 Aug 2021 US
Provisional Applications (1)
Number Date Country
62980867 Feb 2020 US