Cells within a tissue have differences in cell morphology and/or function due to varied analyte levels (e.g., gene and/or protein expression) within the different cells. The specific position of a cell within a tissue (e.g., the cell's position relative to neighboring cells or the cell's position relative to the tissue microenvironment) can affect, e.g., the cell's morphology, differentiation, fate, viability, proliferation, behavior, signaling, and cross-talk with other cells in the tissue.
Spatial heterogeneity has been previously studied using techniques that typically provide data for a handful of analytes in the context of intact tissue or a portion of a tissue (e.g., tissue section), or provide significant analyte data from individual, single cells, but fails to provide information regarding the position of the single cells from the originating biological sample (e.g., tissue).
RNA sequencing libraries generated from tissue samples can pose some challenges. Methods, reaction mixtures, and kits to insert a sequencing adapter directly to the second-strand DNA, thereby adding a second sequencing adapter to a synthesized cDNA previously generated directly on the spatial array would increase efficiency.
Methods for improving the efficiency of adding a sequencing oligonucleotide to a 3′ end of a cDNA generated using a spatial array are desired. The present invention is based on the discovery of an improved method for adding a sequencing oligonucleotide to a 3′ end of a cDNA generated on a spatial array that includes the use of RNAse H or a functional equivalent to produce nicks in the RNA of a RNA:DNA hybrid preceding second strand synthesis followed by end repair, A-tailing, and ligation of a double-stranded adaptor to a blunt-ended double-stranded DNA product. The ligation of a double-stranded adaptor to the blunt-ended double-stranded DNA product is highly efficient, comparative to alternative methods for adaptor incorporation, and contributes to the higher sensitivity of the methods provided herein. The disclosed methods can decrease the hands on time of a researcher in adding a sequencing oligonucleotide to a 3′ end of a cDNA generated on a spatial array (e.g., the methods described herein can be performed within about 12 hours as compared to alternative methods of adaptor incorporation which can take up to one day).
Thus, provided herein are methods of identifying a location of an RNA in a biological sample, the method including: (a) contacting the biological sample with an array including a plurality of capture probes, where a capture probe of the plurality of capture probes includes a capture domain and a spatial barcode; (b) releasing the RNA from the biological sample, where the RNA released from the biological sample is specifically bound by the capture domain of the capture probe; (c) extending a 3′ end of the capture probe using the RNA bound by the capture domain as a template, thereby generating an extended capture probe hybridized to the RNA; (d) generating one or more nick(s) in the RNA hybridized to the extended capture probe and performing random-primed DNA synthesis, thereby replacing the nicked RNA with a second strand DNA molecule hybridized to the extended capture probe; (e) performing end repair on the second strand DNA molecule hybridized to the extended capture probe to generate a blunt-ended double-stranded DNA product; (f) adding a single adenosine (A) nucleotide to the 3′ end of the extended capture probe in the blunt-ended double-stranded DNA product; (g) ligating a double-stranded sequencing adaptor to the double-stranded DNA product; and (h) determining (i) all or a part of the sequence of the RNA or a complement thereof, or (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 RNA in the biological sample.
In some embodiments, generating of the one or more nick(s) in step (d) is performed using RNAse H. In some embodiments, the random-primed DNA synthesis is performed using DNA polymerase I.
In some embodiments, the method includes: (i) the end repair in step (e) is performed using a Klenow fragment and/or T4 DNA polymerase; (ii) step (f) is performed using Taq polymerase; and (iii) step (g) is performed using a kinase, where the kinas is a polynucleotide kinase and a DNA ligase.
In some embodiments, the RNA is a mRNA.
In some embodiments, the method includes: (i) the capture domain is positioned 3′ relative to the spatial barcode in the capture probe and where the capture domain includes a poly(T) sequence; and (ii) the capture probe includes a unique molecular identifier and/or a cleavage domain.
In some embodiments, the method includes between steps (g) and (h), a step of releasing the blunt-ended double-stranded DNA product from the array via cleavage of the cleavage domain.
In some embodiments, the determining in step (h) includes sequencing (i) all or a part of the sequence of the RNA or a complement thereof, or (ii) all or a part of the sequence of the spatial barcode or a complement thereof, where the sequencing is high throughput sequencing, where the high throughput sequencing includes sequencing by ligation or hybridization of labelled probes.
In some embodiments, the method includes creating a library for sequencing from the adaptor-ligated double-stranded DNA product, where the step of creating the library includes amplification, where the amplification includes polymerase chain reaction and the method includes sequencing the library.
In some embodiments, the releasing in step (b), includes permeabilizing the biological sample.
In some embodiments, the array is a slide, where the slide includes beads or wells.
In some embodiments, the biological sample is a tissue sample, a tissue section or a fixed tissue section, and optionally where the fixed tissue section is a formalin-fixed paraffin-embedded tissue section or the tissue section is a fresh, frozen tissue section.
In some embodiments, the method includes one or both of staining and imaging the biological sample.
Also provided herein are reaction mixtures including: an array including a plurality of capture probes, where a capture probe of the plurality includes (i) a spatial barcode and (ii) a capture domain that binds to an RNA; RNAse H; DNA polymerase I; DNA ligase; and a double-stranded sequencing adaptor.
In some embodiments, the reaction mixture includes: (i) the capture domain is positioned 3′ of the spatial barcode in the capture domain; and (ii) the capture probe includes a unique molecular identifier and/or a cleavage domain.
In some embodiments, the reaction mixture includes an RNA from a biological sample and includes one or more of Taq polymerase, a polynucleotide kinase (PNK), a reverse transcriptase, a Klenow fragment, and T4 DNA polymerase.
Also provided herein are kits including: an array including a plurality of capture probes, where a capture probe of the plurality includes (i) a spatial barcode and (ii) a capture domain that binds to an RNA; RNAse H; DNA polymerase I; DNA ligase; and a double-stranded sequencing adaptor.
In some embodiments, the kit includes: (i) the capture domain is positioned 3′ of the spatial barcode in the capture domain; and (ii) the capture probe includes a unique molecular identifier and/or a cleavage domain.
In some embodiments, the array is a slide, where the slide includes beads or wells.
In some embodiments, the kit includes one or more of Taq polymerase, a polynucleotide kinase (PNK), a reverse transcriptase, a Klenow fragment, and T4 DNA polymerase.
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.
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.
Capturing and sequencing as many target analytes from a spatial array is key in determining spatial gene expression in a biological sample. While current methods provide spatial gene expression data certain aspects of the workflow, for example, generating sequence ready libraries from a captured analyte, these methods can be inefficient resulting in a loss of sensitivity and specificity. For example, a certain percentage of the captured analytes can remain unamplified and therefore not sequenced due to workflow inefficiencies resulting in loss of gene expression data. Increasing the efficiencies of such workflows could result in a greater number of captured analytes being amplified and subsequently sequenced, thereby creating a more accurate spatial gene expression picture of a biological sample. The methods disclosed herein provide alternatives to creating sequencing ready libraries from captured analytes on a spatial array by increasing the number of analytes that are processed from library preparation through sequencing from a biological sample, thereby increasing the sensitivity and specificity of a spatial transcriptome array.
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. 2017; and Gupta et al., Nature Biotechnol. 36:1197-1202, 2018; the Visium Spatial Gene Expression Reagent Kits User Guide (e.g., Rev C, dated June 2020), and/or the Visium Spatial Tissue Optimization Reagent Kits User Guide (e.g., Rev C, dated July 2020), both of which are available at the 10× Genomics Support Documentation website, and can be used herein in any combination. Further non-limiting aspects of spatial analysis methodologies and compositions are described herein.
Some general terminology that may be used in this disclosure can be found in Section (I)(b) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Typically, a “barcode” is a label, or identifier, that conveys or is capable of conveying information (e.g., information about an analyte in a sample, a bead, and/or a capture probe). A barcode can be part of an analyte, or independent of an analyte. A barcode can be attached to an analyte. A particular barcode can be unique relative to other barcodes. For the purpose of this disclosure, an “analyte” can include any biological substance, structure, moiety, or component to be analyzed. The term “target” can similarly refer to an analyte of interest.
Analytes can be broadly classified into one of two groups: nucleic acid analytes, and non-nucleic acid analytes. Examples of non-nucleic acid analytes include, but are not limited to, lipids, carbohydrates, peptides, proteins, glycoproteins (N-linked or O-linked), lipoproteins, phosphoproteins, specific phosphorylated or acetylated variants of proteins, amidation variants of proteins, hydroxylation variants of proteins, methylation variants of proteins, ubiquitylation variants of proteins, sulfation variants of proteins, viral proteins (e.g., viral capsid, viral envelope, viral coat, viral accessory, viral glycoproteins, viral spike, etc.), extracellular and intracellular proteins, antibodies, and antigen binding fragments. In some embodiments, the analyte(s) can be localized to subcellular location(s), including, for example, organelles, e.g., mitochondria, Golgi apparatus, endoplasmic reticulum, chloroplasts, endocytic vesicles, exocytic vesicles, vacuoles, lysosomes, etc. In some embodiments, analyte(s) can be peptides or proteins, including without limitation antibodies and enzymes. Additional examples of analytes can be found in Section (I)(c) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. In some embodiments, an analyte can be detected indirectly, such as through detection of an intermediate agent, for example, a ligation product or an analyte capture agent (e.g., an oligonucleotide-conjugated antibody), such as those described herein.
A “biological sample” is typically obtained from the subject for analysis using any of a variety of techniques including, but not limited to, biopsy, surgery, and laser capture microscopy (LCM), and generally includes cells and/or other biological material from the subject. In some embodiments, a biological sample can be a tissue section. In some embodiments, a biological sample can be a fixed and/or stained biological sample (e.g., a fixed and/or stained tissue section). Non-limiting examples of stains include histological stains (e.g., hematoxylin and/or eosin) and immunological stains (e.g., fluorescent stains). In some embodiments, a biological sample (e.g., a fixed and/or stained biological sample) can be imaged. Biological samples are also described in Section (I)(d) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.
In some embodiments, a biological sample is permeabilized with one or more permeabilization reagents. For example, permeabilization of a biological sample can facilitate analyte capture. Exemplary permeabilization agents and conditions are described in Section (I)(d)(ii)(13) or the Exemplary Embodiments Section of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.
Array-based spatial analysis methods involve the transfer of one or more analytes from a biological sample to an array of features on a substrate, where each feature is associated with a unique spatial location on the array. Subsequent analysis of the transferred analytes includes determining the identity of the analytes and the spatial location of the analytes within the biological sample. The spatial location of an analyte within the biological sample is determined based on the feature to which the analyte is bound (e.g., directly or indirectly) on the array, and the feature's relative spatial location within the array.
A “capture probe” refers to any molecule capable of capturing (directly or indirectly) and/or labelling an analyte (e.g., an analyte of interest) in a biological sample. In some embodiments, the capture probe is a nucleic acid or a polypeptide. In some embodiments, the capture probe includes a barcode (e.g., a spatial barcode and/or a unique molecular identifier (UMI)) and a capture domain). In some embodiments, a capture probe can include a cleavage domain and/or a functional domain (e.g., a primer-binding site, such as for next-generation sequencing (NGS)). See, e.g., Section (II)(b) (e.g., subsections (i)-(vi)) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Generation of capture probes can be achieved by any appropriate method, including those described in Section (II)(d)(ii) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.
In some embodiments, more than one analyte type (e.g., nucleic acids and proteins) from a biological sample can be detected (e.g., simultaneously or sequentially) using any appropriate multiplexing technique, such as those described in Section (IV) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.
In some embodiments, detection of one or more analytes (e.g., protein analytes) can be performed using one or more analyte capture agents. As used herein, an “analyte capture agent” refers to an agent that interacts with an analyte (e.g., an analyte in a biological sample) and with a capture probe (e.g., a capture probe attached to a substrate or a feature) to identify the analyte. In some embodiments, the analyte capture agent includes: (i) an analyte binding moiety (e.g., that binds to an analyte), for example, an antibody or antigen-binding fragment thereof; (ii) analyte binding moiety barcode; and (iii) an analyte capture sequence. As used herein, the term “analyte binding moiety barcode” refers to a barcode that is associated with or otherwise identifies the analyte binding moiety. As used herein, the term “analyte capture sequence” refers to a region or moiety configured to hybridize to, bind to, couple to, or otherwise interact with a capture domain of a capture probe. In some cases, an analyte binding moiety barcode (or portion thereof) may be able to be removed (e.g., cleaved) from the analyte capture agent. Additional description of analyte capture agents can be found in Section (II)(b)(ix) of WO 2020/176788 and/or Section (II)(b)(viii) U.S. Patent Application Publication No. 2020/0277663.
There are at least two methods to associate a spatial barcode with one or more neighboring cells, such that the spatial barcode identifies the one or more cells, and/or contents of the one or more cells, as associated with a particular spatial location. One method is to promote analytes or analyte proxies (e.g., intermediate agents) out of a cell and towards a spatially-barcoded array (e.g., including spatially-barcoded capture probes). Another method is to cleave spatially-barcoded capture probes from an array and promote the spatially-barcoded capture probes towards and/or into or onto the biological sample.
The functional sequences can generally be selected for compatibility with any of a variety of different sequencing systems, e.g., Ion Torrent Proton or PGM, Illumina sequencing instruments, PacBio, Oxford Nanopore, etc., and the requirements thereof. In some embodiments, functional sequences can be selected for compatibility with non-commercialized sequencing systems. Examples of such sequencing systems and techniques, for which suitable functional sequences can be used, include (but are not limited to) Ion Torrent Proton or PGM sequencing, Illumina sequencing, PacBio SMRT sequencing, and Oxford Nanopore sequencing. Further, in some embodiments, functional sequences can be selected for compatibility with other sequencing systems, including non-commercialized sequencing systems.
In some embodiments, the spatial barcode 105 and functional sequences 104 is common to all of the probes attached to a given feature. In some embodiments, the UMI sequence 106 of a capture probe attached to a given feature is different from the UMI sequence of a different capture probe attached to the given feature.
In some cases, capture probes may be configured to prime, replicate, and consequently yield optionally barcoded extension products from a template (e.g., a DNA or RNA template, such as an analyte or an intermediate agent (e.g., a ligation product or an analyte capture agent), or a portion thereof), or derivatives thereof (see, e.g., Section (II)(b)(vii) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663 regarding extended capture probes). In some cases, capture probes may be configured to form ligation products with a template (e.g., a DNA or RNA template, such as an analyte or an intermediate agent, or portion thereof), thereby creating ligations products that serve as proxies for a template.
As used herein, an “extended capture probe” refers to a capture probe having additional nucleotides added to the terminus (e.g., 3′ or 5′ end) of the capture probe thereby extending the overall length of the capture probe. For example, an “extended 3′ end” indicates additional nucleotides were added to the most 3′ nucleotide of the capture probe to extend the length of the capture probe, for example, by polymerization reactions used to extend nucleic acid molecules including templated polymerization catalyzed by a polymerase (e.g., a DNA polymerase or a reverse transcriptase). In some embodiments, extending the capture probe includes adding to a 3′ end of a capture probe a nucleic acid sequence that is complementary to a nucleic acid sequence of an analyte or intermediate agent specifically bound to the capture domain of the capture probe. In some embodiments, the capture probe is extended using reverse transcription. In some embodiments, the capture probe is extended using one or more DNA polymerases. The extended capture probes include the sequence of the capture probe and the sequence of the spatial barcode of the capture probe.
In some embodiments, extended capture probes are amplified (e.g., in bulk solution or on the array) to yield quantities that are sufficient for downstream analysis, e.g., via DNA sequencing. In some embodiments, extended capture probes (e.g., DNA molecules) act as templates for an amplification reaction (e.g., a polymerase chain reaction).
Additional variants of spatial analysis methods, including in some embodiments, an imaging step, are described in Section (II)(a) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Analysis of captured analytes (and/or intermediate agents or portions thereof), for example, including sample removal, extension of capture probes, sequencing (e.g., of a cleaved extended capture probe and/or a cDNA molecule complementary to an extended capture probe), sequencing on the array (e.g., using, for example, in situ hybridization or in situ ligation approaches), temporal analysis, and/or proximity capture, is described in Section (II)(g) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Some quality control measures are described in Section (II)(h) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.
Spatial information can provide information of biological and/or medical importance. For example, the methods and compositions described herein can allow for: identification of one or more biomarkers (e.g., diagnostic, prognostic, and/or for determination of efficacy of a treatment) of a disease or disorder; identification of a candidate drug target for treatment of a disease or disorder; identification (e.g., diagnosis) of a subject as having a disease or disorder; identification of stage and/or prognosis of a disease or disorder in a subject; identification of a subject as having an increased likelihood of developing a disease or disorder; monitoring of progression of a disease or disorder in a subject; determination of efficacy of a treatment of a disease or disorder in a subject; identification of a patient subpopulation for which a treatment is effective for a disease or disorder; modification of a treatment of a subject with a disease or disorder; selection of a subject for participation in a clinical trial; and/or selection of a treatment for a subject with a disease or disorder.
Spatial information can provide information of biological importance. For example, the methods and compositions described herein can allow for: identification of transcriptome and/or proteome expression profiles (e.g., in healthy and/or diseased tissue); identification of multiple analyte types in close proximity (e.g., nearest neighbor analysis); determination of up- and/or down-regulated genes and/or proteins in diseased tissue; characterization of tumor microenvironments; characterization of tumor immune responses; characterization of cells types and their co-localization in tissue; and identification of genetic variants within tissues (e.g., based on gene and/or protein expression profiles associated with specific disease or disorder biomarkers).
Typically, for spatial array-based methods, a substrate functions as a support for direct or indirect attachment of capture probes to features of the array. A “feature” is an entity that acts as a support or repository for various molecular entities used in spatial analysis. In some embodiments, some or all of the features in an array are functionalized for analyte capture. Exemplary substrates are described in Section (II)(c) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663. Exemplary features and geometric attributes of an array can be found in Sections (II)(d)(i), (II)(d)(iii), and (II)(d)(iv) of WO 2020/176788 and/or U.S. Patent Application Publication No. 2020/0277663.
Generally, analytes and/or intermediate agents (or portions thereof) can be captured when contacting a biological sample with a substrate including capture probes (e.g., a substrate with capture probes embedded, spotted, printed, fabricated on the substrate, or a substrate with features (e.g., beads, wells) comprising capture probes). As used herein, “contact,” “contacted,” and/or “contacting,” a biological sample with a substrate refers to any contact (e.g., direct or indirect) such that capture probes can interact (e.g., bind covalently or non-covalently (e.g., hybridize)) with analytes from the biological sample. Capture can be achieved actively (e.g., using electrophoresis) or passively (e.g., using diffusion). Analyte capture is further described in Section (II)(e) of WO 2020/176788 and/or U.S. Patent Application Publication No. 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.
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.
Methods for Increasing the Efficiency of Library Preparation of Nucleic Acids from a Spatial Array
Capturing and sequencing as many target analytes from a spatial array is key in determining spatial gene expression in a biological sample. While current methods provide spatial gene expression data certain aspects of the workflow, for example, generating sequence ready libraries from a captured analyte, can be inefficient resulting in a loss of sensitivity and specificity. For example, a certain percentage of the captured analytes can remain unamplified and therefore not sequenced due to workflow inefficiencies resulting in loss of gene expression data. Increasing the efficiencies of such workflows could result in a greater number of captured analytes being amplified and subsequently sequenced thereby creating a more accurate spatial gene expression picture of a biological sample. The methods disclosed herein provide alternatives to creating sequencing ready libraries from captured analytes on a spatial array by increasing the number of analytes that are processed from library preparation through sequencing from a biological sample, thereby increasing the sensitivity and specificity of a spatial transcriptome array.
The present invention is based on the discovery of an improved method for adding a sequencing oligonucleotide to a 3′ end of a cDNA generated on a spatial array that includes the use of RNAse H or a functional equivalent to produce nicks in the RNA of a RNA:DNA hybrid preceding second strand synthesis followed by end repair, A-tailing, and ligation of a double-stranded adaptor to a blunt-ended double-stranded DNA product. The ligation of a double-stranded adaptor to the blunt-ended double-stranded DNA product is highly efficient, relative to alternative methods for adaptor incorporation, and contributes to the higher sensitivity of the methods provided herein. The disclosed methods can decrease the hands-on time of a researcher in adding a sequencing oligonucleotide to a 3′ end of a cDNA generated on a spatial array (e.g., the methods described herein can be performed within about 12 hours as compared to alternative methods of adaptor incorporation which can take up to one day).
A schematic showing an exemplary embodiment of the methods provided herein is shown in
Thus, provided herein are methods of identifying a location of an RNA (e.g., mRNA or any of the other exemplary types of RNAs described herein or known in the art) in a biological sample (e.g., any of the exemplary biological samples described herein) that include: (a) contacting the biological sample with an array (e.g., any of the exemplary arrays described herein) comprising a plurality of capture probes, wherein a capture probe of the plurality of capture probes comprises a capture domain (e.g., any of the exemplary capture domains described herein) and a spatial barcode; (b) releasing the RNA from the biological sample, where the RNA released from the biological sample is bound by the capture domain of the capture probe; (c) extending a 3′ end of the capture probe using the RNA bound by the capture domain as a template, thereby generating an extended capture probe hybridized to the RNA; (d) generating one or more nick(s) in the RNA hybridized to the extended capture probe and performing random-primed DNA synthesis (e.g., using DNA polymerase I, and optionally, a DNA ligase), thereby replacing the nicked RNA with a second strand DNA molecule hybridized to the extended capture probe; (e) performing end repair on the second strand DNA molecule hybridized to the extended capture probe to generate a blunt-ended double-stranded DNA product (e.g., using a Klenow fragment and/or a T4 DNA polymerase); (f) adding a single adenosine (A) nucleotide to the 3′ end of the extended capture probe in the blunt-ended double-stranded DNA product (e.g., using Taq polymerase); (g) ligating a double-stranded sequencing adaptor to the double-stranded DNA product (e.g., using a kinase, e.g., a polynucleotide kinase, and a DNA ligase); and (h) determining (i) all or a part of the sequence of the RNA or a complement thereof, or (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 RNA in the biological sample.
A double-stranded adaptor sequencing adaptor can be any suitable double-stranded sequencing adaptor that can be used to perform any of the exemplary sequencing methods described herein. In some embodiments, the double-stranded sequencing adaptor can include an Illumina sequencing adaptor sequence. In some embodiments, the double-stranded sequencing adaptor can include a first and a second strand that each can be about 15 to 45 nucleotides (e.g., about 15 to about 45 nucleotides, about 15 to about 40 nucleotides, about 15 to about 35 nucleotides, about 15 to about 30 nucleotides, about 15 to about 25 nucleotides, or about 15 to about 20 nucleotides, about 20 to about 45 nucleotides, about 20 to about 40 nucleotides, about to about 35 nucleotides, about 20 to about 30 nucleotides, about 20 to about 25 nucleotides, about 25 to about 45 nucleotides, about 25 to about 40 nucleotides, about 25 to about 35 nucleotides, about 25 to about 30 nucleotides, about 30 to about 45 nucleotides, about 30 to about 40 nucleotides, about 30 to about 35 nucleotides, about 35 to about 45 nucleotides, about to about 40 nucleotides, or about 40 to about 45 nucleotides) long. In some embodiments, the double-stranded sequencing adaptor can include a universal sequence.
In some embodiments of any of the methods described herein, the capture domain comprises a poly(T) sequence. In some embodiments, the capture domain does not comprise a poly(T) sequence. In some embodiments, the capture domain comprises a sequence that is substantially complementary to a contiguous sequence present in the RNA. The capture domain can be about 5 to about 40 nucleotides (e.g., about 5 to about 35 nucleotides, about 5 to about 30 nucleotides, about 5 to about 25 nucleotides, about 5 to about 20 nucleotides, about 5 to about 15 nucleotides, about 5 to about 10 nucleotides, about 10 to about 40 nucleotides, about 10 to about nucleotides, about 10 to about 30 nucleotides, about 10 to about 25 nucleotides, about 10 to about 20 nucleotides, about 10 to about 15 nucleotides, about 15 to about 40 nucleotides, about to about 35 nucleotides, about 15 to about 30 nucleotides, about 15 to about 25 nucleotides, about 15 to about 20 nucleotides, about 20 to about 40 nucleotides, about 20 to about 35 nucleotides, about 20 to about 30 nucleotides, about 20 to about 25 nucleotides, about 25 to about 40 nucleotides, about 25 to about 35 nucleotides, about 25 to about 30 nucleotides, about to about 40 nucleotides, about 30 to about 35 nucleotides, or about 35 to about 40 nucleotides) in length.
In some embodiments, the capture domain is positioned 3′ relative to the spatial barcode in the capture probe. In some embodiments of any of the methods provided herein, the capture probe further includes a unique molecular identifier, a cleavage domain (e.g., any of the exemplary cleavage domains described herein), one or more functional domains (e.g., a sequencing primer domain), or combinations thereof.
In some embodiments, the step of extending the 3′ end of the capture probe using the RNA bound by the capture domain as a template includes the use of a reverse transcriptase. In some embodiments, the step of generating the one or more nick(s) in the RNA hybridized to the extended capture probe is performed using RNAse H or a functional equivalent thereof. In some embodiments, the random-primed DNA synthesis is performed using DNA polymerase I, and optionally, a ligase (e.g., DNA ligase). In some embodiments, the end repair on the second strand DNA molecule hybridized to the extended capture probe to generate a blunt-ended double-stranded DNA product, is performed using a Klenow fragment and/or T4 DNA polymerase. In some embodiments, the step of adding a single adenosine (A) nucleotide to the 3′ end of the extended capture probe in the blunt-ended double-stranded DNA product is performed using Taq polymerase. In some embodiments, the step of ligating a double-stranded sequencing adaptor to the double-stranded DNA product is performed using a kinase (e.g., a polynucleotide kinase) and a DNA ligase.
In some embodiments, the capture probe further comprises a cleavage domain. Some embodiments of any of the methods described herein can further include, between the step of ligating the double-stranded sequencing adaptor to the double-stranded DNA product and the step of determining (i) all or a part of the sequence of the RNA or a complement thereof, or (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 RNA in the biological sample, a step of releasing the blunt-ended double-stranded DNA product from the array via cleavage of the cleavage domain.
In some embodiments of any of the methods described herein, the step of releasing the RNA from the biological sample comprises permeabilizing the biological sample (e.g., using any one or more of any of the permeabilization agents described herein and/or any of the physical conditions (e.g., heating) described herein.
In some embodiments of any of the methods described herein, the plurality of capture probes are affixed (i.e., attached) to an array. In some embodiments of any of the methods described herein, the array is a slide (e.g., a slide comprising beads or a slide comprising wells (e.g., microwells)). An array can also have one or more of any of the exemplary characteristics of arrays described herein.
In some embodiments, the step of determining (i) all or a part of the sequence of the RNA or a complement thereof, or (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 RNA in the biological sample comprises sequencing (i) all or a part of the sequence of the RNA or a complement thereof, or (ii) all or a part of the sequence of the spatial barcode or a complement thereof. In some embodiments, the sequencing can be performed using any of the exemplary sequencing methods described herein (e.g., high throughput sequencing). In some embodiments of any of the methods described herein, the RNA is mRNA. In some embodiments, the RNA is any of the other exemplary types of RNA described herein or known in the art.
In some embodiments of any of the methods described herein, the biological sample can be any of the exemplary biological samples described herein. For example, a biological sample can be a tissue sample or a tissue section. For example, the biological sample can be a fixed tissue section (e.g., a formalin-fixed paraffin-embedded (FFPE) tissue section, acetone fixed tissues section, methanol fixed tissue section, or a paraformaldehyde fixed tissue section) or a fresh, frozen tissue section.
Some embodiments of any of the methods described herein can further include one or both of staining and imaging the biological sample (e.g., using any of the exemplary stains and staining methods described herein and using any of the exemplary imaging methods described herein). In some embodiments of any of the methods described herein, the biological sample was previously stained and/or imaged (e.g., before the releasing in step (b)). In some embodiments, the staining can include the use of an optical label as described herein, including, but not limited to, fluorescent, radioactive, chemiluminescent, calorimetric, or colorimetric detectable labels. In some embodiments, staining can include the use of a fluorescent antibody directed to a target analyte (e.g., cell surface or intracellular proteins) in the biological sample. In some embodiments, staining can include the use of a chemical stain, such as hematoxylin and eosin (H&E) or periodic acid-schiff (PAS), or immunofluorescence (IF) techniques as known in the art. In some embodiments, significant time (e.g., days, months, or years) can elapse between staining and/or imaging the biological sample and performing any of the methods described herein.
Reaction Mixtures and Kits
Also provided herein are reaction mixtures that include: an array comprising a plurality of capture probes affixed thereon, wherein a capture probe of the plurality comprises (i) a spatial barcode and (ii) a capture domain that binds specifically to an RNA; RNAse H; DNA polymerase I; DNA ligase; and a double-stranded sequencing adaptor.
Also provided herein are kits that include: an array comprising a plurality of capture probes, where a capture probe of the plurality comprises (i) a spatial barcode and (ii) a capture domain that binds specifically to an RNA; RNAse H; DNA polymerase I; DNA ligase; and a double-stranded sequencing adaptor.
In some embodiments of any of the reaction mixtures or kits described herein, the capture domain can be any of the capture domains described herein. In some embodiments, the capture domain can comprise a poly(T) sequence. In some embodiments, the capture domain does not comprise a poly(T) sequence. In some embodiments, the capture domain comprises a sequence that is substantially complementary to a contiguous sequence present in the RNA. The capture domain can be about 5 to about 40 nucleotides (e.g., or any of the subranges of this range described herein) in length.
In some embodiments, the capture domain is positioned 3′ relative to the spatial barcode in the capture probe. In some embodiments of any of the reaction mixtures or kits provided herein, the capture probe further includes a unique molecular identifier, a cleavage domain (e.g., any of the exemplary cleavage domains described herein), a sequencing primer domain, a universal sequence, or combinations thereof. In some embodiments, the sequencing primer domain is positioned at the 5′ end relative to the capture domain, the spatial barcode, the unique molecular identifier, the cleavage domain, the universal sequence, or any combination thereof. In some embodiments, the sequencing primer is affixed to the substrate.
In some embodiments, the sequencing primer domain is positioned 5′ relative to the capture domain and the spatial barcode. In some embodiments, a functional domain (e.g., a sequencing primer) domain is positioned 5′ relative to the capture domain, the spatial barcode, and the unique molecular identifier. In some embodiments, the sequencing primer domain is positioned 3′ relative to the cleavage domain. In some embodiments, the sequencing primer domain is on the 5′ end that is affixed to the substrate.
In some embodiments of any of the reaction mixtures or kits described herein, the plurality of capture probes are affixed (i.e., attached) to an array. In some embodiments of any of the reaction mixtures or kits described herein, the array is a slide (e.g., a slide comprising beads or a slide comprising wells (e.g., microwells)). An array can also have one or more of any of the exemplary characteristics of arrays described herein.
A double-stranded adaptor sequencing adaptor can be any suitable double-stranded sequencing adaptor that can be used to perform any of the exemplary sequencing methods described herein. In some embodiments, the double-stranded sequencing adaptor can include an Illumina sequencing adaptor sequence. In some embodiments, the double-stranded sequencing adaptor can include a first and a second strand that each can be about 15 to 45 nucleotides (e.g., any of the subranges of this range described herein) long. In some embodiments, the double-stranded sequencing adaptor can include a universal sequence.
In some embodiments of any of the reaction mixtures described herein, the reaction mixture can include an RNA from a biological sample (e.g., an mRNA or any of the other types of RNA described herein or known in the art).
Some embodiments of any of the reaction mixtures or kits described herein further include one or both of Taq polymerase and a polynucleotide kinase (PNK). Some embodiments of any of the reaction mixtures or kits described herein further include a reverse transcriptase. Some embodiments of any of the reaction mixtures or kits described herein further include one or both of Klenow fragment and T4 DNA polymerase.
Some embodiments of any of the kits or reaction mixtures described herein can further include one or more permeabilization reagents (e.g., one or more of any of the permeabilization reagents described herein), such as for example, a protease.
Some embodiments of any of the kits described herein can further include a staining agent. In some embodiments, a staining agent can include an optical label, e.g., a fluorescent, a radioactive, a chemiluminescent, a calorimetric, or a colorimetric detectable label. In some embodiments, a staining agent can be a fluorescent antibody directed to a target analyte (e.g., cell surface or intracellular proteins). In some embodiments, a staining agent can be a chemical stain, such as hematoxylin and eosin (H&E) or periodic acid-schiff (PAS).
Some embodiments of any of the kits described herein can further include instructions for performing any of the methods described herein.
Embodiment 1 is a method of identifying a location of an RNA in a biological sample, the method comprising: (a) contacting the biological sample with an array comprising a plurality of capture probes, wherein a capture probe of the plurality of capture probes comprises a capture domain and a spatial barcode; (b) releasing the RNA from the biological sample, wherein the RNA released from the biological sample is specifically bound by the capture domain of the capture probe; (c) extending a 3′ end of the capture probe using the RNA specifically bound by the capture domain as a template, thereby generating an extended capture probe hybridized to the RNA; (d) generating one or more nick(s) in the RNA hybridized to the extended capture probe and performing random-primed DNA synthesis, thereby replacing the nicked RNA with a second strand DNA molecule hybridized to the extended capture probe; (e) performing end repair on the second strand DNA molecule hybridized to the extended capture probe to generate a blunt-ended double-stranded DNA product; (f) adding a single adenosine (A) nucleotide to the 3′ end of the extended capture probe in the blunt-ended double-stranded DNA product; (g) ligating a double-stranded sequencing adaptor to the double-stranded DNA product; and (h) determining (i) all or a part of the sequence of the RNA or a complement thereof, or (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 RNA in the biological sample.
Embodiment 2 is the method of embodiment 1, wherein the generating of the one or more nick(s) in step (d) is performed using RNAse H.
Embodiment 3 is the method of embodiment 1, wherein the random-primed DNA synthesis is performed using DNA polymerase I.
Embodiment 4 is the method of any one of embodiments 1-3, wherein the end repair in step (e) is performed using a Klenow fragment and/or T4 DNA polymerase.
Embodiment 5 is the method of any one of embodiments 1-4, wherein step (f) is performed using Taq polymerase.
Embodiment 6 is the method of any one of embodiments 1-5, wherein step (g) is performed using a kinase and a DNA ligase.
Embodiment 7 is the method of embodiment 6, wherein the kinase is a polynucleotide kinase (PNK).
Embodiment 8 is the method of any one of embodiments 1-7, wherein the RNA is a mRNA.
Embodiment 9 is the method of embodiment 8, wherein the capture domain comprises a poly(T) sequence.
Embodiment 10 is the method of any one of embodiments 1-9, wherein the capture domain is positioned 3′ relative to the spatial barcode in the capture probe.
Embodiment 11 is the method of any one of embodiments 1-10, wherein the capture probe further comprises a unique molecular identifier.
Embodiment 12 is the method of any one of embodiments 1-11, wherein the capture probe further comprises a cleavage domain.
Embodiment 13 is the method of embodiment 12, wherein the method further comprises, between steps (g) and (h), a step of releasing the blunt-ended double-stranded DNA product from the array via cleavage of the cleavage domain.
Embodiment 14 is the method of any one of embodiments 1-13, wherein the determining in step (h) comprises sequencing (i) all or a part of the sequence of the RNA or a complement thereof, or (ii) all or a part of the sequence of the spatial barcode or a complement thereof.
Embodiment 15 is the method of embodiment 14, wherein the sequencing is high throughput sequencing.
Embodiment 16 is the method of embodiment 15, wherein the high throughput sequencing comprises sequencing by ligation.
Embodiment 17 is the method of embodiment 15, wherein the high throughput sequencing comprises hybridization of labelled probes.
Embodiment 18 is the method of any one of embodiments 1-14, wherein the method further comprises creating a library for sequencing from the adaptor-ligated double-stranded DNA product.
Embodiment 19 is the method of embodiment 18, wherein the step of creating the library comprises amplification.
Embodiment 20 is the method of embodiment 19, wherein the amplification comprises polymerase chain reaction.
Embodiment 21 is the method of embodiment 20, wherein the method further comprises sequencing the library.
Embodiment 22 is the method of any one of embodiments 1-21, wherein the releasing in step (b), comprises permeabilizing the biological sample.
Embodiment 23 is the method of any one of embodiments 1-22, wherein the array is a slide.
Embodiment 24 is the method of embodiment 23, wherein the slide comprises beads.
Embodiment 25 is the method of embodiment 23, wherein the slide comprises wells.
Embodiment 26 is the method of any one of embodiments 1-25, wherein the biological sample is a tissue sample.
Embodiment 27 is the method of embodiment 26, wherein the tissue sample is a tissue section.
Embodiment 28 is the method of embodiment 27, wherein the tissue section is a fixed tissue section.
Embodiment 29 is the method of embodiment 28, wherein the fixed tissue section is a formalin-fixed paraffin-embedded tissue section.
Embodiment 30 is the method of embodiment 27, wherein the tissue section is a fresh, frozen tissue section.
Embodiment 31 is the method of any one of embodiments 1-30, wherein the method further comprising one or both of staining and imaging the biological sample.
Embodiment 32 is a reaction mixture comprising: an array comprising a plurality of capture probes, wherein a capture probe of the plurality comprises (i) a spatial barcode and (ii) a capture domain that binds specifically to an RNA; RNAse H; DNA polymerase I; DNA ligase; and a double-stranded sequencing adaptor.
Embodiment 33 is the reaction mixture of embodiment 32, wherein the capture domain is positioned 3′ of the spatial barcode in the capture domain.
Embodiment 34 is the reaction mixture of embodiment 32 or 33, wherein the capture probe further comprises a unique molecular identifier.
Embodiment 35 is the reaction mixture of any one of embodiments 32-34, wherein the capture probe further comprises a cleavage domain.
Embodiment 36 is the reaction mixture of any one of embodiments 32-35, wherein the reaction mixture further comprises an RNA from a biological sample.
Embodiment 37 is the reaction mixture of any one of embodiments 32-36, wherein the array is a slide.
Embodiment 38 is the reaction mixture of embodiment 37, wherein the slide comprises beads.
Embodiment 39 is the reaction mixture of embodiment 37, wherein the array comprises wells.
Embodiment 40 is the reaction mixture of any one of embodiments 32-39, wherein the reaction mixture further comprises one or both of Taq polymerase and a polynucleotide kinase (PNK).
Embodiment 41 is the reaction mixture of any one of embodiments 32-40, wherein the reaction mixture further comprises reverse transcriptase.
Embodiment 42 is the reaction mixture of any one of embodiments 32-41, wherein the reaction mixture further comprises one or both of Klenow fragment and T4 DNA polymerase.
Embodiment 43 is a kit comprising: an array comprising a plurality of capture probes, wherein a capture probe of the plurality comprises (i) a spatial barcode and (ii) a capture domain that binds specifically to an RNA; RNAse H; DNA polymerase I; DNA ligase; and a double-stranded sequencing adaptor.
Embodiment 44 is the kit of embodiment 43, wherein the capture domain is positioned 3′ of the spatial barcode in the capture domain.
Embodiment 45 is the kit of embodiment 43 or 44, wherein the capture probe further comprises a unique molecular identifier.
Embodiment 46 is the kit of any one of embodiments 43-45, wherein the capture probe further comprises a cleavage domain.
Embodiment 47 is the kit of any one of embodiments 43-46, wherein the array is a slide.
Embodiment 48 is the kit of embodiment 47, wherein the slide comprises beads.
Embodiment 49 is the kit of embodiment 47, wherein the array comprises wells.
Embodiment 50 is the kit of any one of embodiments 43-49, wherein the kit further comprises one or both of Taq polymerase and a polynucleotide kinase (PNK).
Embodiment 51 is the kit of any one of embodiments 43-50, wherein the kit further comprises reverse transcriptase.
Embodiment 52 is the kit of any one of embodiments 43-51, wherein the kit further comprises one or both of Klenow fragment and T4 DNA polymerase.
This application is a continuation of U.S. patent application Ser. No. 17/368,276, filed Jul. 6, 2021 (issued as U.S. Pat. No. 11,761,038, which claims priority to U.S. Provisional Patent Application No. 63/048,627, filed on Jul. 6, 2020, the contents of each of which are incorporated herein by reference in their entirety.
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 |
4988617 | Landegren et al. | Jan 1991 | 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 |
7499806 | Kermani et al. | Mar 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 |
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 |
9005935 | Belyaev | 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 et al. | 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 |
9790476 | Gloeckner 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 |
10266876 | Cai et al. | Apr 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 |
11384386 | Chee | Jul 2022 | B2 |
11390912 | Frisen et al. | Jul 2022 | B2 |
11401545 | Chee | Aug 2022 | B2 |
11407992 | Dadhwal | Aug 2022 | B2 |
11408029 | Katiraee et al. | Aug 2022 | B2 |
11434524 | Ramachandran Iyer et al. | Sep 2022 | B2 |
11479809 | Frisen et al. | Oct 2022 | B2 |
11479810 | Chee | Oct 2022 | B1 |
11492612 | Dadhwal | Nov 2022 | B1 |
11505828 | Chell et al. | Nov 2022 | B2 |
11512308 | Gallant et al. | Nov 2022 | B2 |
11519022 | Chee | Dec 2022 | B2 |
11519033 | Schnall-Levin et al. | Dec 2022 | B2 |
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 |
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 |
11845979 | Engblom et al. | Dec 2023 | B2 |
11859178 | Gallant et al. | Jan 2024 | B2 |
11866767 | Uytingco et al. | Jan 2024 | B2 |
11866770 | Chee | Jan 2024 | B2 |
11873482 | Kim et al. | Jan 2024 | 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 |
20020058250 | Firth | 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 | Fan et al. | Jun 2011 | A1 |
20110245101 | Chee et al. | Oct 2011 | A1 |
20110245111 | Chee | Oct 2011 | A1 |
20110287435 | Grunenwald et al. | Nov 2011 | A1 |
20120021930 | Schoen et al. | Jan 2012 | 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 |
20120220479 | Ericsson 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 |
20120301925 | Belyaev | 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 |
20130065768 | Zheng et al. | Mar 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 |
20130302801 | Asbury et al. | Nov 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 |
20160194692 | Gore et al. | Jul 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 |
20170159109 | Zheng et al. | Jun 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 |
20170283860 | Kool et al. | Oct 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 |
20190233880 | Mir | 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 |
20190360043 | Pham 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 | 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 |
20220241780 | Tentori et al. | Aug 2022 | A1 |
20220267844 | Ramachandran Iyer et al. | Aug 2022 | A1 |
20220282329 | Chell et al. | Sep 2022 | A1 |
20220290217 | Frenz et al. | Sep 2022 | A1 |
20220290219 | Chee | Sep 2022 | A1 |
20220298560 | Frisen et al. | Sep 2022 | A1 |
20220325325 | Chee et al. | Oct 2022 | A1 |
20220326251 | Uytingco et al. | Oct 2022 | A1 |
20220333171 | Chee | Oct 2022 | A1 |
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 |
20230416807 | Chee | Jan 2023 | A1 |
20230416808 | Sukovich et al. | 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 |
20230392204 | Chell et al. | Dec 2023 | A1 |
20230393071 | Bava | Dec 2023 | A1 |
20230407404 | Baumgartner et al. | Dec 2023 | A1 |
20230416850 | Singh et al. | Dec 2023 | A1 |
20240002931 | Bava | Jan 2024 | A1 |
20240011081 | Chee | Jan 2024 | A1 |
20240011090 | Chew et al. | Jan 2024 | A1 |
20240018572 | Mignardi | Jan 2024 | A1 |
20240018575 | Gallant et al. | Jan 2024 | A1 |
20240018589 | Schnall-Levin et al. | Jan 2024 | A1 |
20240026445 | Ramachandran Iyer et al. | Jan 2024 | A1 |
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 |
1929039 | Jun 2008 | EP |
2002017 | Dec 2008 | EP |
2292788 | Mar 2011 | EP |
2302070 | Mar 2011 | EP |
2580351 | Apr 2013 | 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 1998044151 | Oct 1998 | WO |
WO 2000017390 | Mar 2000 | WO |
WO 2000063437 | Oct 2000 | WO |
WO 2001006012 | Jan 2001 | WO |
WO 2001009363 | Feb 2001 | WO |
WO 2001012862 | Feb 2001 | WO |
WO 2001042796 | Jun 2001 | WO |
WO 2001046402 | Jun 2001 | WO |
WO 2001059161 | Aug 2001 | WO |
WO 2001090415 | Nov 2001 | WO |
WO 2001096608 | Dec 2001 | WO |
WO 2002040874 | May 2002 | 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 2006137733 | Dec 2006 | WO |
WO 2007037678 | Apr 2007 | 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 2008093098 | Aug 2008 | WO |
WO 2009032167 | Mar 2009 | WO |
WO 2009036525 | Mar 2009 | WO |
WO 2009152928 | Dec 2009 | WO |
WO 2010019826 | Feb 2010 | WO |
WO 2010027870 | Mar 2010 | WO |
WO 2010126614 | Nov 2010 | WO |
WO 2010127186 | Nov 2010 | WO |
WO 2011008502 | Jan 2011 | WO |
WO 2011062933 | May 2011 | WO |
WO 2011068088 | Jun 2011 | WO |
WO 2011127006 | Oct 2011 | WO |
WO 2011155833 | Dec 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 2013150082 | Oct 2013 | WO |
WO 2013150083 | Oct 2013 | WO |
WO 2014044724 | Mar 2014 | WO |
WO 2014060483 | Apr 2014 | WO |
WO 2014071361 | May 2014 | WO |
WO 2014130576 | Aug 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 2015069374 | May 2015 | WO |
WO 2015161173 | Oct 2015 | WO |
WO 2016077763 | May 2016 | WO |
WO 2016138496 | Sep 2016 | WO |
WO 2016138500 | Sep 2016 | WO |
WO 2016162309 | Oct 2016 | WO |
WO 2016166128 | Oct 2016 | WO |
WO 2016168825 | Oct 2016 | WO |
WO 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 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 2019165318 | Aug 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 2020198071 | Oct 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 2021067514 | Apr 2021 | WO |
WO 2021091611 | May 2021 | WO |
WO 2021092433 | May 2021 | WO |
WO 2021097255 | May 2021 | WO |
WO 2021102003 | May 2021 | WO |
WO 2021102005 | May 2021 | WO |
WO 2021102039 | May 2021 | WO |
WO 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 2021142233 | 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 2021252747 | Dec 2021 | WO |
WO 2021263111 | Dec 2021 | WO |
WO 2022025965 | Feb 2022 | WO |
WO 2022060798 | Mar 2022 | WO |
WO 2022060953 | Mar 2022 | WO |
WO 2022061152 | Mar 2022 | WO |
WO 2022087273 | Apr 2022 | WO |
WO 2022098810 | May 2022 | WO |
WO 2022099037 | May 2022 | WO |
WO 2022103712 | May 2022 | WO |
WO 2022109181 | May 2022 | WO |
WO 2022132645 | Jun 2022 | WO |
WO 2022140028 | Jun 2022 | WO |
WO 2022147005 | Jul 2022 | WO |
WO 2022147296 | Jul 2022 | WO |
WO 2022164615 | Aug 2022 | WO |
WO 2022178267 | Aug 2022 | WO |
WO 2022198068 | Sep 2022 | WO |
WO 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 2022226372 | Dec 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 2023215552 | Nov 2023 | WO |
WO 2023225519 | Nov 2023 | WO |
WO 2023229988 | Nov 2023 | WO |
WO 2023250077 | Dec 2023 | WO |
WO 2024015578 | Jan 2024 | WO |
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. |
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. |
U.S. Appl. No. 63/033,348, filed Jun. 2, 2020, Bent. |
[No Author Listed], “Chromium Next GEM Single Cell 3′ Reagent Kits v3.1—User Guide,” 10x Genomics, Document No. CG000204, Nov. 2019, 58 pages. |
[No Author Listed], “Chromium Next GEM Single Cell 3′ Reagent Kits v3.1 (Dual Index)—User Guide,” 10x Genomics, Mar. 2021, Document No. CG000315, 61 pages. |
[No Author Listed], “HuSNP Mapping Assay User's Manual,” Affymetrix Part No. 90094 (Affymetrix, Santa Clara, Calif.), GeneChip, 2000, 104 pages. |
[No Author Listed], “Microarray technologies have excellent possibilities in genomics-related researches,” Science Tools From Amersham Pharmacia Biotech, 1998, 3(4): 8 pages (with English Translation). |
[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—Tissue Optimization—User Guide,” Jul. 2020, retrieved on May 25, 2021, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/5UJrN0cH17rEk0UXwd19It/e54d99fb08a8f1500aba503005a04a56/CG000238_VisiumSpatialTissueOptimizationUserGuide_RevD.pdf>, 42 pages. |
10xGenomics.com, [online], “Visium Spatial Gene Expression Reagent Kits—Tissue Optimization,” Nov. 2019, retrieved on Jan. 25, 2022, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/4q03w6959AJFxffSw5lee9/6a2ac61cf6388a72564eeb96bc294967/CG000238_VisiumSpatialTissueOptimizationUserGuide_Rev_A.pdf>, 46 pages. |
10xGenomics.com, [online], “Visium Spatial Gene Expression Reagent Kits—Tissue Optimization,” Oct. 2020, retrieved on Dec. 28, 2021, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/5UJrN0cH17rEk0UXwd19It/e54d99fb08a8f1500aba503005a04a56/CG000238_VisiumSpatialTissueOptimizationUserGuide_RevD.pdf>, 43 pages. |
10xGenomics.com, [online], “Visium Spatial Gene Expression Reagent Kits—User Guide,” Jun. 2020, retrieved on May 25, 2021, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/3GGIfH3RWpd1bFVhalpexR/8baa08d9007157592b65b2cdc7130990/CG000239_VisiumSpatialGeneExpression_UserGuide_RevD.pdf>, 69 pages. |
10xGenomics.com, [online], “Visium Spatial Gene Expression Reagent Kits—User Guide,” Oct. 2020, retrieved on Dec. 28, 2021, retrieved from URL<https://assets.ctfassets.net/an68im79xiti/3GGIfH3RWpd1bFVhalpexR/8baa08d9007157592b65b2cdc7130990/CG000239_ VisiumSpatialGeneExpression_UserGuide_RevD.pdf>, 70 pages. |
Adessi et al., “Solid phase DNA amplification: characterisation of primer attachment and amplification mechanisms,” Nucl. Acids Res., 2000, 28(20):E87, 8 pages. |
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. |
Alam, “Proximity Ligation Assay (PLA),” Curr Protoc Immunol., Nov. 2018, 123(1):e58, 8 pages. |
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. |
Amidzadeh et al., “Assessment of different permeabilization methods of minimizing damage to the adherent cells for detection of intracellular RNA by flow cytometry,” Avicenna J Med Biotechnol., Jan. 2014, 6(1):38-46. |
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. |
Appella, “Non-natural nucleic acids for synthetic biology,” Current Opinion in Chemical Biology, Dec. 2009, 13(5-6): 687-696. |
Aran et al., “XCell: digitally portraying the tissue cellular heterogeneity landscape,” Genome Biol., Nov. 2017, 18(1):220, 14 pages. |
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. |
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. |
Asp et al., “Spatially Resolved Transcriptomes-Next Generation Tools for Tissue Exploration,” Bioessays, Oct. 2020, 42(10):e1900221, 16 pages. |
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. |
Balakrishnan et al., “Flap endonuclease 1,” Annu Rev Biochem., Jun. 2013, 82:119-138. |
Baner et al., “Signal amplification of padlock probes by rolling circle replication,” Nucleic Acids Res., 1998, 26(22):5073-5078. |
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. |
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. |
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. |
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. |
Bell, “A simple way to treat PCR products prior to sequencing using ExoSAP-IT,” Biotechniques, 2008, 44(6):834, 1 page. |
Belton et al., “Hi-C: A comprehensive technique to capture the conformation of genomes,” Methods, Nov. 2012, 58(3):268-276, 16 pages. |
Bentley et al., “Accurate whole human genome sequencing using reversible terminator chemistry,” Nature, 2008, 456(7218):53-59. |
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. |
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. |
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. |
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. |
Blair et al., “Microarray temperature optimization using hybridization kinetics,” Methods Mol Biol., 2009, 529:171-96. |
Blanchard et al., “High-density oligonucleotide arrays,” Biosensors & Bioelectronics, 1996, 11(6-7):687-690. |
Blanco et al., “A practical approach to FRET-based PNA fluorescence in situ hybridization,” Methods, Dec. 2010, 52(4):343-51. |
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. |
Bolotin et al., “MiXCR: software for comprehensive adaptive immunity profiling,” Nat Methods., May 2015, 12(5):380-1. |
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). |
Boulé et al., “Terminal deoxynucleotidyl transferase indiscriminately incorporates ribonucleotides and deoxyribonucleotides,” J Biol Chem., Aug. 2001, 276(33):31388-93. |
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. |
Bunt et al., “FRET from single to multiplexed signaling events,” Biophys Rev. Apr. 2017, 9(2): 119-129. |
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. |
Burgess, “Spatial transcriptomics coming of age,” Nat Rev Genet., Jun. 2019, 20(6):317, 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. |
Caliari et al., “A practical guide to hydrogels for cell culture,” Nat Methods., Apr. 2016, 13(5):405-14. |
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. “Arrayed profiling of multiple glycans on whole living cell surfaces.” Analytical chemistry, Oct. 15, 2013, 85(22):11153-11158. |
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., “Efficient in situ barcode sequencing using padlock probe-based BaristaSeq,” Nucleic Acids Res., 2018, 46(4): e22, 11 pages. |
Chen et al., “Expansion microscopy,” Science, 2015, 347(6221):543-548. |
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. |
Chen et al., “Nanoscale imaging of RNA with expansion microscopy,” Nat Methods, Aug. 2016, 13(8):679-84. |
Chen et al., “Parallel single nucleotide polymorphism genotyping by surface invasive cleavage with universal detection,” Anal Chem., Apr. 2005, 77(8):2400-5. |
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. |
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. |
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. |
Choi et al., “Multiplexed detection of mRNA using porosity-tuned hydrogel microparticles,” Analytical chemistry, Sep. 28, 2012, 84(21):9370-9378. |
Chrisey et al., “Covalent attachment of synthetic DNA to self-assembled monolayer films,” Nucleic Acids Res., Aug. 1996, 24(15):3031-9. |
Ciaccio et al., “Systems analysis of EGF receptor signaling dynamics with microwestern arrays,” Nat Methods, Feb. 2010, 7(2):148-55. |
Codeluppi et al., “Spatial organization of the somatosensory cortex revealed by osmFISH,” Nature Methods, Nov. 2018, 15:932-935. |
Constantine et al., “Use of genechip high-density oligonucleotide arrays for gene expression monitoring,” Life Science 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. |
Cruz et al., “Methylation in cell-free DNA for early cancer detection,” Ann Oncol., Jun. 2018, 29(6):1351-1353. |
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. |
Czarnik, “Encoding methods for combinatorial chemistry,” Curr Opin Chem Biol., Jun. 1997, 1(1):60-6. |
Dahl et al., “Circle-to-circle amplification for precise and sensitive DNA analysis,” Proc. Natl. Acad. Sci., 2004, 101(13):4548-4553. |
Dalma-Weiszhausz et al., “The affymetrix GeneChip platform: an overview,” Methods Enzymol., 2006, 410:3-28. |
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. |
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. |
Deng et al., “Spatial Epigenome Sequencing at Tissue Scale and Cellular Level,” BioRxiv, Mar. 2021, 40 pages. |
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. |
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. |
Evers et al., “The effect of formaldehyde fixation on RNA: optimization of formaldehyde adduct removal,” J Mol Diagn., May 2011, 13(3):282-8. |
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. |
Fan et al., “Illumina Universal Bead Arrays,” Methods in Enzymology, 2006, 410:57-73. |
Faruqi et al., “High-throughput genotyping of single nucleotide polymorphisms with rolling circle amplification,” BMC Genomics, Aug. 2001, 2:4, 10 pages. |
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., “Continuous Polony Gels for Tissue Mapping with High Resolution and RNA Capture Efficiency,” bioRxiv, 2021, 20 pages. |
Fu et al., “Counting individual DNA molecules by the stochastic attachment of diverse labels,” PNAS, 2011, 108(22):9026-9031. |
Fu et al., “Repeat subtraction-mediated sequence capture from a complex genome,” Plant J., Jun. 2010, 62(5):898-909. |
Fullwood et al., “Next-generation DNA sequencing of paired-end tags (PET) for transcriptome and genome analyses,” Genome Res., 2009, 19(4):521-532. |
Ganguli et al., “Pixelated spatial gene expression analysis from tissue,” Nat Commun., Jan. 2018, 9(1):202, 9 pages. |
Gansauge et al., “Single-stranded DNA library preparation from highly degraded DNA using T4 DNA ligase,” Nucleic Acids Res., Jun. 2017, 45(10):e79, 10 pages. |
Gao et al., “A highly homogeneous expansion microscopy polymer composed of tetrahedron-like monomers,” bioRxiv, Oct. 22, 2019, 23 pages (Preprint). |
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. |
Gerard et al., “Excess dNTPs minimize RNA hydrolysis during reverse transcription,” Biotechniques, Nov. 2002, 33(5):984, 986, 988, 990. |
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. |
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. |
Goh et al., “Highly Specific Multiplexed RNA Imaging in Tissues With Split-FISH,” Nat Methods, Jun. 15, 2020, 17(7):689-693, 21 pages. |
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. |
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. |
Goransson et al., “A single molecule array for digital targeted molecular analyses,” Nucleic Acids Res., Nov. 25, 2009, 37(1):e7, 9 pages. |
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. |
Grokhovsky, “Specificity of DNA cleavage by ultrasound,” Molecular Biology, 2006, 40(2):276-283. |
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. |
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. |
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. |
Hafner et al., “Identification of microRNAs and other small regulatory RNAs using cDNA library sequencing,” Methods, Jan. 2008, 44(1):3-12. |
Hahnke et al., “Striptease on glass: validation of an improved stripping procedure for in situ microarrays,” J Biotechnol., Jan. 2007, 128(1):1-13. |
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. |
Hanauer et al., “Separation of nanoparticles by gel electrophoresis according to size and shape,” Nano Lett., Sep. 2007, 7(9):2881-5. |
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. |
Hayes et al., “Electrophoresis of proteins and nucleic acids: I-Theory,” BMJ, Sep. 1989, 299(6703):843-6. |
He et al., “In situ synthesis of protein arrays,” Current Opinion in Biotechnology, 2008, 19(1):4-9. |
He 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. |
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. |
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. |
Ho et al., “Characterization of an ATP-Dependent DNA Ligase Encoded by Chlorella Virus PBCV-1,” Journal of Virology, Mar. 1997, 71(3):1931-1937. |
Hoffman et al., “Formaldehyde crosslinking: a tool for the study of chromatin complexes,” J Biol Chem., Oct. 2015, 290(44):26404-11. |
Hsuih et al., “Novel, Ligation-Dependent PCR Assay for Detection of Hepatitis C Virus in Serum,” Journal of Clinical Microbiology, Mar. 1996, 34(3):501-507. |
Hu et al., “High reproducibility using sodium hydroxide-stripped long oligonucleotide DNA microarrays,” Biotechniques, Jan. 2005, 38(1):121-4. |
Hughes et al., “Microfluidic Western blotting,” PNAS, Dec. 2012, 109(52):21450-21455. |
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. |
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. |
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. |
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. |
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. |
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. |
Ke et al., “In situ sequencing for RNA analysis in preserved tissue and cells,” Nat Methods., Sep. 2013, Supplementary Materials, 29 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. |
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. |
Kumar et al., “Template-directed oligonucleotide strand ligation, covalent intramolecular DNA circularization and catenation using click chemistry,” J Am Chem Soc., May 2007, 129(21):6859-64. |
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. |
Lahiani et al., “Enabling Histopathological Annotations on Immunofluorescent Images through Virtualization of Hematoxylin and Eosin,” J Pathol Inform., Feb. 2018, 9:1, 8 pages. |
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. |
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. |
Leriche et al., “Cleavable linkers in chemical biology,” Bioorganic & Medicinal Chemistry, 2012, 20:571-582. |
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. |
Li et al., “A photocleavable fluorescent nucleotide for DNA sequencing and analysis,” Proc. Natl. Acad. Sci., 2003, 100(2):414-419. |
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. |
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. |
Liu et al., “High-Spatial-Resolution Multi-Omics 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. |
Lizardi et al., “Mutation detection and single-molecule counting using isothermal rolling-circle amplification,” Nat. Genet., 1998, 19(3):225-232. |
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. |
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. |
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. |
Lyck et al., “Immunohistochemical markers for quantitative studies of neurons and glia in human neocortex,” J Histochem Cytochem, 2008, 56(3):201-21. |
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. |
Ma et al., “Isothermal amplification method for next-generation sequencing,” PNAS, Aug. 12, 2013, 110(35):14320-14323. |
MacBeath et al., “Printing proteins as microarrays for high-throughput function determination,” Science, Sep. 2000, 289(5485):1760-1763. |
MacIntyre, “Unmasking antigens for immunohistochemistry.,” Br J Biomed Sci., 2001, 58(3):190-6. |
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. |
Marx, “Method of the Year: spatially resolved transcriptomics,” Nature Methods, 2021, 18(1):9-14. |
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. |
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 profiling of proteins and RNA in fixed tissue,” Nat Biotechnol, May 2020, 38(5):586-599. |
Metzker, “Sequencing technologies—the next generation,” Nature Reviews Genetics, 2010, 11(1):31-46. |
Miele et al., “Mapping cis- and trans-chromatin interaction networks using chromosome conformation capture (3C),” Methods Mol Biol., 2009, 464:105-21. |
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. |
Miller et al., “Basic concepts of microarrays and potential applications in clinical microbiology,” Clinical Microbiology Reviews, 2009, 22(4):611-633. |
Miller et al., “Chapter 11—Solid and Suspension Microarrays for Microbial Diagnostics,” Methods in Microbiology, 2015, 42:395-431. |
Miner et al., “Molecular barcodes detect redundancy and contamination in hairpin-bisulfite PCR,” Nucleic Acids Res., Sep. 2004, 32(17):e135, 4 pages. |
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. |
Miura et al., “Highly efficient single-stranded DNA ligation technique improves low-input whole-genome bisulfite sequencing by post-bisulfite adaptor tagging,” Nucleic Acids Res., Sep. 2019, 47(15):e85, 10 pages. |
Mizusawa et al., “A bacteriophage lambda vector for cloning with BamHI and Sau3A,” Gene, 1982, 20(3):317-322. |
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. |
Morlan et al., “Selective depletion of rRNA enables whole transcriptome profiling of archival fixed tissue, ” PLoS One, Aug. 2012, 7(8):e42882, 8 pages. |
Motea et al., “Terminal deoxynucleotidyl transferase: the story of a misguided DNA polymerase,” Biochim Biophys Acta., May 2010, 1804(5):1151-66. |
Mulder et al., “CapTCR-seq: hybrid capture for T-cell receptor repertoire profiling,” Blood Advances, Dec. 2018, 2(23):3506-3514. |
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. |
Nallur et al., “Signal amplification by rolling circle amplification on DNA microarrays,” Nucleic Acids Res., Dec. 1, 2001, 29(23):e118, 9 pages. |
Nandakumar et al., “How an RNA Ligase Discriminates RNA versus DNA Damage,” Molecular Cell, 2004, 16:211-221. |
Nandakumar et al., “RNA Substrate Specificity and Structure-guided Mutational Analysis of Bacteriophage T4 RNA Ligase 2,” Journal of Biological Chemistry, Jul. 2004, 279(30):31337-31347. |
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. |
Niedringhaus et al., “Landscape of next-generation sequencing technologies,” Anal Chem., Jun. 2011, 83(12):4327-41. |
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. |
Nilsson et al., “RNA-templated DNA ligation for transcript analysis,” Nucleic Acids Res., Jan. 2001, 29(2):578-81. |
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. |
Oksuz et al., “Systematic evaluation of chromosome conformation capture assays,” Nature Methods, Sep. 2021, 18:1046-1055. |
Olivier, “The Invader assay for SNP genotyping,” Mutat. Res., Jun. 2005, 573(1-2):103-110. |
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. |
Ozsolak et al., “Digital transcriptome profiling from attomole-level RNA samples,” Genome Res., Apr. 2010, 20(4):519-25. |
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. |
Passow et al., “RNAlater and flash freezing storage methods nonrandomly influence observed gene expression in RNAseq experiments,” bioRxiv, Jul. 2018, 28 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. |
Pemov et al., “DNA analysis with multiplex microarray-enhanced PCR,” Nucl. Acids Res., Jan. 2005, 33(2):e11, 9 pages. |
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. |
Perler et al., “Intervening sequences in an Archaea DNA polymerase gen,” Proc Natl Acad Sci USA, Jun. 1992, 89(12):5577-5581. |
Perocchi et al., “Antisense artifacts in transcriptome microarray experiments are resolved by actinomycin D,” Nucleic Acids Res., 2007, 35(19):e128, 7 pages. |
Petterson et al., “Generations of sequencing technologies,” Genomics, 2009, 93(2):105-111. |
Picelli et al., “Full-length RNA-seq from single cells using Smart-seq2,” Nat Protoc., Jan. 2014, 9(1):171-81. |
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. |
Plongthongkum et al., “Advances in the profiling of DNA modifications: cytosine methylation and beyond,” Nature Reviews Genetics, Aug. 2014, 15(10):647-661. |
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. |
Porreca et al., “Polony DNA sequencing,” Curr Protoc Mol Biol., Nov. 2006, Chapter 7, Unit 7.8, p. 7.8.1-7.8.22. |
U.S. Appl. No. 61/267,363, filed Dec. 7, 2009, 33 pages. |
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. |
Raab et al., “Human tRNA genes function as chromatin insulators,” EMBO J., Jan. 2012, 31(2):330-50. |
Raj et al., “Imaging individual mRNA molecules using multiple singly labeled probes,” Nature Methods, Oct. 2008, 5(10):877-879, 9 pages. |
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. |
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. |
Rodriques et al., “Slide-seq: A scalable technology for measuring genome-wide expression at high spatial resolution, ” Science, 2019, 363(6434):1463-1467. |
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. |
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. |
Salmén et al., “Barcoded solid-phase RNA capture for Spatial Transcriptomics profiling in mammalian tissue sections,” Nature Protocols, Oct. 2018, 13(11):2501-2534. |
Saxonov et al., “10x Genomics, Mastering Biology to Advance Human Health,” PowerPoint, 10x, 2020, 41 pages. |
Schena et al., “Quantitative monitoring of gene expression patterns with a complementary DNA microarray,” Science, Oct. 1995, 270(5235):467-470. |
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. |
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. |
Schweitzer et al., “Multiplexed protein profiling on microarrays by rolling-circle amplification,” Nature Biotechnology, Apr. 2002, 20(4):359-365. |
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. |
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. |
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. |
Skene et al., “An efficient targeted nuclease strategy for high-resolution mapping of DNA binding sites,” Elife, Jan. 2017, 6:e21856, 35 pages. |
Slomovic et al., “Addition of poly(A) and poly(A)-rich tails during RNA degradation in the cytoplasm of human cells,” Proc Natl Acad Sci USA, Apr. 2010, 107(16):7407-12. |
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. |
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, Jul. 2016, 353(6294):78-82. |
Stahl et al., “Visualization and analysis of gene expression in tissue sections by spatial transcriptomics,” Supplementary Materials, Science, Jul. 2016, 353(6294):78-82, 41 pages. |
Stimpson et al., “Real-time detection of DNA hybridization and melting on oligonucleotide arrays by using optical wave guides,” Proc Natl Acad Sci USA, Jul. 1995, 92(14):6379-83. |
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. |
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. |
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. |
Svensson et al., “SpatialDE: identification of spatially variable genes,” Nature Methods, May 2018, 15:343-346, 15 pages. |
Takei et al., “Integrated Spatial Genomics Reveals Global Architecture of Single Nuclei,” Nature, Jan. 27, 2021, 590(7845):344-350, 53 pages. |
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. |
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. |
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. |
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. |
Twyman et al., “Techniques Patents for SNP Genotyping,” Pharmacogenomics, Jan. 2003, 4(1):67-79. |
Ulery et al., “Biomedical Applications of Biodegradable Polymers,” J Polym Sci B Polym Phys., Jun. 2011, 49(12):832-864. |
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. |
Vandenbroucke et al., “Quantification of splice variants using real-time PCR,” Nucleic Acids Research, 2001, 29(13):e68, 7 pages. |
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. |
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. |
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,” Nat Methods, Oct. 2019, 16(10):987-990. |
Vickovic et al., “SM-Omics: An automated Platform for High-Throughput Spatial Multi-Omics,” bioRxiv, Oct. 2020, 40 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 et al., “Imaging-based pooled CRISPR screening reveals regulators of IncRNA localization,” Proc Natl Acad Sci USA, May 2019, 116(22):10842-10851. |
Wang et al., “Optimization of Process Conditions for Infected Animal Tissues by Alkaline Hydrolysis Technology,” Procedia Environmental Sciences, 2016, 31:366-374. |
Wang et al., “Tagmentation-based whole-genome bisulfite sequencing,” Nature Protocols, Oct. 2013, 8(10):2022-2032. |
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. |
Wiedmann et al., “Ligase chain reaction (LCR)—overview and applications,” PCR Methods Appl., Feb. 1994, 3(4):S51-64. |
Wilson et al., “New transposon delivery plasmids for insertional mutagenesis in Bacillus anthracis,” J Microbiol Methods, Dec. 2007, 71(3):332-5. |
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. |
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. |
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. |
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. |
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. |
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. |
Yeakley et al., “Profiling alternative splicing on fiber-optic arrays,” Nature biotechnology, 2002, 20:353-358. |
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. |
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. |
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., “Genome-wide open chromatin regions and their effects on the regulation of silk protein genes in Bombyx mori,” Sci Rep., Oct. 2017, 7(1):12919, 9 pages. |
Zhang et al., “Multiplex ligation-dependent probe amplification (MLPA) for ultrasensitive multiplexed microRNA detection using ribonucleotide-modified DNA probest,” Chem. Commun., 2013, 49:10013-10015. |
Zhao et al., “Isothermal Amplification of Nucleic Acids,” Chemical Reviews, Nov. 2015, 115(22):12491-12545. |
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. |
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. |
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. |
Schmidl et al., “ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors,” Nature Methods, Oct. 2015, 12:963-965. |
Number | Date | Country | |
---|---|---|---|
20230383344 A1 | Nov 2023 | US |
Number | Date | Country | |
---|---|---|---|
63048627 | Jul 2020 | US |
Number | Date | Country | |
---|---|---|---|
Parent | 17368276 | Jul 2021 | US |
Child | 18448618 | US |