US20190300945A1 - Spatially Encoded Biological Assays - Google Patents

Spatially Encoded Biological Assays Download PDF

Info

Publication number
US20190300945A1
US20190300945A1 US16/437,726 US201916437726A US2019300945A1 US 20190300945 A1 US20190300945 A1 US 20190300945A1 US 201916437726 A US201916437726 A US 201916437726A US 2019300945 A1 US2019300945 A1 US 2019300945A1
Authority
US
United States
Prior art keywords
tissue sample
assay
peptide
biological
probes
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/437,726
Inventor
Mark S. Chee
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Prognosys Biosciences Inc
Original Assignee
Prognosys Biosciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US13/080,616 external-priority patent/US9371598B2/en
Priority claimed from US16/414,213 external-priority patent/US10787701B2/en
Priority to US16/437,726 priority Critical patent/US20190300945A1/en
Application filed by Prognosys Biosciences Inc filed Critical Prognosys Biosciences Inc
Assigned to PROGNOSYS BIOSCIENCES, INC. reassignment PROGNOSYS BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEE, MARK S.
Priority to US16/443,771 priority patent/US20190309355A1/en
Publication of US20190300945A1 publication Critical patent/US20190300945A1/en
Priority to US16/660,234 priority patent/US10662468B2/en
Priority to US16/670,603 priority patent/US10612079B2/en
Priority to US16/734,216 priority patent/US10619196B1/en
Priority to US16/837,924 priority patent/US10983113B2/en
Priority to US16/913,672 priority patent/US11733238B2/en
Priority to US18/341,373 priority patent/US20230358733A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B60/00Apparatus specially adapted for use in combinatorial chemistry or with libraries
    • C40B60/04Integrated apparatus specially adapted for both screening libraries and identifying library members
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • C12Q1/6837Enzymatic or biochemical coupling of nucleic acids to a solid phase using probe arrays or probe chips
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/543Immunoassay; Biospecific binding assay; Materials therefor with an insoluble carrier for immobilising immunochemicals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6804Nucleic acid analysis using immunogens
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6809Methods for determination or identification of nucleic acids involving differential detection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6827Hybridisation assays for detection of mutation or polymorphism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6834Enzymatic or biochemical coupling of nucleic acids to a solid phase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6841In situ hybridisation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • C12Q1/6874Methods for sequencing involving nucleic acid arrays, e.g. sequencing by hybridisation
    • CCHEMISTRY; METALLURGY
    • C40COMBINATORIAL TECHNOLOGY
    • C40BCOMBINATORIAL CHEMISTRY; LIBRARIES, e.g. CHEMICAL LIBRARIES
    • C40B30/00Methods of screening libraries
    • C40B30/04Methods of screening libraries by measuring the ability to specifically bind a target molecule, e.g. antibody-antigen binding, receptor-ligand binding
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/5308Immunoassay; Biospecific binding assay; Materials therefor for analytes not provided for elsewhere, e.g. nucleic acids, uric acid, worms, mites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6845Methods of identifying protein-protein interactions in protein mixtures
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6848Methods of protein analysis involving mass spectrometry
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2458/00Labels used in chemical analysis of biological material
    • G01N2458/10Oligonucleotides as tagging agents for labelling antibodies
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2458/00Labels used in chemical analysis of biological material
    • G01N2458/40Rare earth chelates

Definitions

  • This invention relates to assays of biological molecules, and more particularly to assays for determining spatial distributions of a large number of biological molecules in a solid sample simultaneously.
  • RNA sequences per sample include microarrays (see Shi, et al., Nature Biotechnology, 24(9):1151-61 (2006); and Slonim and Yanai, Plos Computational Biology, 5(10):e1000543 (2009)); serial analysis of gene expression (SAGE) (see Velculescu, et al, Science, 270(5235):484-87 (1995)), high-throughput implementations of qPCR (see Spurgeon, et al., Plos ONE, 3(2):e1662 (2008)) and in situ PCR (see Nuovo, Genome Res., 4:151-67 (1995)).
  • SAGE serial analysis of gene expression
  • qPCR see Spurgeon, et al., Plos ONE, 3(2):e1662 (2008)
  • in situ PCR see Nuovo, Genome Res., 4:151-67 (1995)
  • the invention encompasses assay systems that provide high-resolution spatial maps of biological activity in tissues.
  • the assay system comprises an assay capable of high levels of multiplexing where encoded probes are provided to a biological sample in defined spatial patterns; instrumentation capable of controlled delivery of reagents according to the spatial patterns; and a decoding scheme providing a readout that is digital in nature.
  • the present invention provides the ability to look at many biological targets in many locations, providing the resolution of in situ hybridization with the highly- parallel data analysis of sequencing.
  • the invention provides an assay system to determine spatial patterns of abundance or activity or both of multiple biological targets at multiple sites in a sample, where the assay system performs the following steps: providing a sample affixed to a support; delivering encoded probes for the multiple biological targets to the multiple sites in the sample in a known spatial pattern, where each encoded probe comprises a probe region that may interact with the biological targets and a coding tag that identifies a location of the site to which the encoded probe was delivered; allowing the encoded probes to interact with the biological targets; separating encoded probes that interact with the biological targets from encoded probes that do not interact with the biological targets; determining all or a portion of a sequence of the encoded probes, and associating the abundance or activity or both of the multiple biological targets to the locations of the sites in the sample.
  • the biological targets comprise nucleic acids and the encoded probes are oligonucleotides, and in some aspects, there are two encoded probes for each of the multiple nucleic acid targets.
  • the multiple biological targets comprise proteins, the probe regions of the encoding probes are proteins and the coding tags comprise oligonucleotides.
  • the multiple biological targets comprise enzymes.
  • the probe regions of the encoded probes comprise antibodies, aptamers or small molecules.
  • the assay system further comprise an amplification step between the separating step and the determining step.
  • the determining step is performed by nucleic acid sequencing, and in preferred aspects, the sequencing is high- throughput digital nucleic acid sequencing.
  • the product of the multiple biological targets being assayed and the multiple sites in the sample is greater than 20, in some aspects product of the multiple biological targets being assayed and the multiple sites in the sample is greater than 50, in some aspects the product of the multiple biological targets being assayed and the multiple sites in the sample is greater than 75, 100, 150, 500, 750, 1,000, 5,000, 10,000, 25,000, 50,000, 100,000, 500,000, or 1,000,000 or more.
  • sequence of at least fifty thousand encoding probes are determined in parallel, in other aspects the sequence of at least one hundred thousand encoding probes are determined in parallel, in some aspects the sequence of at least five hundred thousand encoding probes are determined in parallel, and in some aspects the sequence of at least one million, ten million, one hundred million, one billion, ten billion, one hundred billion or more encoding probes are determined in parallel.
  • the known spatial pattern is determined by histological features of the sample.
  • software programmed hardware performs at least two steps of the delivering step, the separation step, the determining step and the associating step.
  • the probe regions of the encoded probes are proteins and the separating step is accomplished by encoded probes that interact with the biological targets being captured by an affinity capture agent.
  • the probe regions of the encoding probes are nucleic acids and the separating step is accomplished by a washing of the sample.
  • an assay system to determine spatial patterns of abundance or activity or both of multiple nucleic acid targets at multiple sites in a sample, where the assay system performs the following steps: providing a sample affixed to a support; delivering oligonucleotide probes for multiple nucleic acid targets to the multiple sites in the sample in a known spatial pattern; allowing the oligonucleotide probes to hybridize with the nucleic acid targets; washing unhybridized encoded oligonucleotide probes from the sample; delivering one or more encoding agents to locations of the multiple sites in the sample according to a known spatial pattern, where the combination of encoding agents delivered to each site is different; coupling the encoding agents and the oligonucleotide probes to form encoded probes; determining all or a portion of a sequence of the encoded probes using high-throughput sequencing, and associating the abundance or activity or both of multiple biological targets to the locations of multiple sites in the sample.
  • an assay system to determine spatial patterns of abundance or activity or both of multiple protein targets at multiple sites in a sample, where the assay system performs the following steps: providing a sample affixed to a support; delivering encoded probes for the multiple protein targets to the multiple sites in the sample in a known spatial pattern, where each encoded probe comprises a protein probe region that may interact with the protein targets and a coding tag that identifies a location of the site to which the encoded probe was delivered and the protein probe region of the encoding probe of which the coding tag is part; allowing the encoded probes to interact with the protein targets; separating encoded probes that interact with the protein targets from encoded probes that do not interact with the protein targets; determining all or a portion of a sequence of the encoded probes by high throughput sequencing, and associating the abundance or activity or both of the multiple protein targets to the locations of the multiple sites in the sample.
  • an assay system to determine spatial patterns of abundance or activity or both of multiple biological targets at multiple sites in a sample, where the assay system performs the following steps: providing a sample affixed to a support; delivering encoded probes for the multiple biological targets to the multiple sites in the sample in a known spatial pattern, where each encoded probe comprises a probe region that may interact with the biological targets and a coding tag that identifies a location of the site to which the encoded probe was delivered and identifies the biological target; allowing the encoded probes to interact with the biological targets; determining all or a portion of a sequence of the encoded probes, and associating the abundance or activity or both of the multiple biological targets to the locations of the sites in the sample.
  • the assay system of the invention can utilize various detection mechanisms, based on the molecules to be detected and the reagents needed for such detection system. Exemplary methods that can be used with the assay systems of the invention are described in more detail below.
  • FIG. 1 provides a simplified overview of the assay system of the present invention.
  • FIG. 2 provides a simplified overview of one embodiment of the assay system of the present invention for detecting nucleic acids.
  • FIG. 3 is a representational depiction of one embodiment of the assay overviewed in FIG. 2 .
  • FIG. 4A-C illustrates a general mechanism for one embodiment of a combinatorial encoding scheme of the assay systems of the invention.
  • FIG. 4A shows two target-specific/encoding oligonucleotide constructs specifically bound to a target nucleic acid of interest in a sample.
  • FIG. 4B shows a scheme for delivering twenty different coding tags, a1 through a10 and b1 through b10, to a sample to form a 10 ⁇ 10 coding tag grid.
  • FIG. 4C shows a tissue section sample to which the coding tags are delivered, forming the coding tag grid in the sample.
  • FIG. 5 provides a simplified, specific example of the embodiment of a combinatorial encoding scheme shown in FIG. 4 .
  • antibody as used herein is intended to refer to an entire immunoglobulin or antibody or any functional fragment of an immunoglobulin molecule which is capable of specific binding to an antigen (antibodies and antigens are “binding partners” as defined herein). “Antibody” as used herein is meant to include the entire antibody as well as any antibody fragments capable of binding the antigen or antigenic fragment of interest. Examples of such peptides include complete antibody molecules, antibody fragments, such as Fab, F(ab′)2, CDRS, VL, VH, and any other portion of an antibody which is capable of specifically binding to an antigen.
  • Antibodies for assays of the invention are immunoreactive or immunospecific for, and therefore specifically and selectively bind to, proteins either detected (i.e., biological targets) or used for detection (i.e., probes) in the assays of the invention.
  • binding agent refers to any agent that specifically binds to a biological molecule of interest
  • “Complementary” or “substantially complementary” refers to the hybridization or base pairing or the formation of a duplex between nucleotides or nucleic acids, such as, for instance, between the two strands of a double-stranded DNA molecule or between an oligonucleotide primer and a primer binding site on a single-stranded nucleic acid.
  • Complementary nucleotides are, generally, A and T (or A and U), or C and G.
  • Two single-stranded RNA or DNA molecules are said to be substantially complementary when the nucleotides of one strand, optimally aligned and compared and with appropriate nucleotide insertions or deletions, pair with at least about 80% of the other strand, usually at least about 90% to about 95%, and even about 98% to about 100%).
  • Hybridization refers to the process in which two single-stranded polynucleotides bind non-covalently to form a stable double-stranded polynucleotide, The resulting (usually) double-stranded polynucleotide is a “hybrid” or “duplex,” “Hybridization conditions” will typically include salt concentrations of approximately less than 1M, often less than about 500 mM and may be less than about 200 mM.
  • a “hybridization buffer” is a buffered salt solution such as 5% SSPE, or other such buffers known in the art.
  • Hybridization temperatures can be as low as 5° C., but are typically greater than 22° C., and more typically greater than about 30° C., and typically in excess of 37° C.
  • Hybridizations are often performed under stringent conditions, i.e., conditions under which a primer will hybridize to its target subsequence but will not hybridize to the other, non-complementary sequences.
  • Stringent conditions are sequence-dependent and are different in different circumstances. For example, longer fragments may require higher hybridization temperatures for specific hybridization than short fragments.
  • the combination of parameters is more important than the absolute measure of any one parameter alone.
  • Generally stringent conditions are selected to be about 5° C. lower than the Tm for the specific sequence at a defined ionic strength and pH.
  • Exemplary stringent conditions include a salt concentration of at least 0.01 M to no more than 1M sodium ion concentration (or other salt) at a pH of about 7.0 to about 8.3 and a temperature of at least 25° C.
  • 5 ⁇ SSPE 750 mM NaCl, 50 mM sodium phosphate, 5 mM EDTA at pH 7.4
  • a temperature of approximately 30° C. are suitable for allele-specific hybridizations, though a suitable temperature depends on the length and/or GC content of the region hybridized.
  • “Ligation” means to form a covalent bond or linkage between the termini of two or more nucleic acids, e.g., oligonucleotides and/or polynucleotides, in a template-driven reaction.
  • the nature of the bond or linkage may vary widely and the ligation may be carried out enzymatically or chemically.
  • ligations are usually carried out enzymatically to form a phosphodiester linkage between a 5′ carbon terminal nucleotide of one oligonucleotide with a 3′ carbon of another nucleotide.
  • Nucleic acid refers generally to at least two nucleotides covalently linked together.
  • a nucleic acid generally will contain phosphodiester bonds, although in some cases nucleic acid analogs may be included that have alternative backbones such as phosphoramidite, phosphorodithioate, or methylphophoroamidite linkages; or peptide nucleic acid backbones and linkages.
  • Other analog nucleic acids include those with bicyclic structures including locked nucleic acids, positive backbones, non-ionic backbones and non-ribose backbones. Modifications of the ribose-phosphate backbone may be done to increase the stability of the molecules; for example, PNA:DNA hybrids can exhibit higher stability in some environments.
  • Primer means an oligonucleotide, either natural or synthetic, that is capable, upon forming a duplex with a polynucleotide template, of acting as a point of initiation of nucleic acid synthesis and being extended from its 3′ end along the template so that an extended duplex is formed.
  • the sequence of nucleotides added during the extension process is determined by the sequence of the template polynucleotide. Primers usually are extended by a DNA polymerase.
  • SNP single nucleotide polymorphism
  • SNPs are found across the genome; much of the genetic variation between individuals is due to variation at SNP loci, and often this genetic variation results in phenotypic variation between individuals.
  • SNPs for use in the present invention and their respective alleles may be derived from any number of sources, such as public databases (U.C.
  • a biallelic genetic marker is one that has two polymorphic forms, or alleles.
  • biallelic genetic marker that is associated with a trait
  • the allele that is more abundant in the genetic composition of a case group as compared to a control group is termed the “associated allele,” and the other allele may be referred to as the “unassociated allele.”
  • the associated allele the allele that is more abundant in the genetic composition of a case group as compared to a control group
  • the other allele may be referred to as the “unassociated allele.”
  • associated allele e.g., a disease or drug response
  • Other biallelic polymorphisms that may be used with the methods presented herein include, but are not limited to multinucleotide changes, insertions, deletions, and translocations.
  • references to DNA herein may include genomic DNA, mitochondrial DNA, episomal DNA, and/or derivatives of DNA such as amplicons, RNA transcripts, cDNA, DNA analogs, etc.
  • the polymorphic loci that are screened in an association study may be in a diploid or a haploid state and, ideally, would be from sites across the genome.
  • binding partner e.g., protein, nucleic acid, antibody or other affinity capture agent, etc.
  • binding partner e.g., protein, nucleic acid, antibody or other affinity capture agent, etc.
  • specific binding will be at least three times the standard deviation of the background signal.
  • the binding partner binds to its particular “target” molecule and does not bind in a significant amount to other molecules present in the sample.
  • Sequence determination means determination of information relating to the nucleotide base sequence of a nucleic acid. Such information may include the identification or determination of partial as well as full sequence information of the nucleic acid. Sequence information may be determined “with varying degrees of statistical reliability or confidence. In one aspect, the term includes the determination of the identity and ordering of a plurality of contiguous nucleotides in a nucleic acid, “High throughput digital sequencing” or “next generation sequencing” means sequence determination using methods that determine many (typically thousands to billions) of nucleic acid sequences in an intrinsically parallel manner, i.e.
  • DNA templates are prepared for sequencing not one at a time, but in a bulk process, and where many sequences are read out preferably in parallel, or alternatively using an ultra-high throughput serial process that itself may be parallelized.
  • Such methods include but are not limited to pyrosequencing (for example, as commercialized by 454 Life Sciences, Inc., Branford, Conn.); sequencing by ligation (for example, as commercialized in the SOLiDTM technology, Life Technology, Inc., Carlsbad, Calif.); sequencing by synthesis using modified nucleotides (such as commercialized in TruSeqTM and HiSeqTM technology by Illumina, Inc., San Diego, Calif., HeliScopeTM by Helicos Biosciences Corporation, Cambridge, Mass., and PacBio RS by Pacific Biosciences of California, Inc., Menlo Park, Calif.), sequencing by ion detection technologies (Ion Torrent, Inc., South San Francisco, Calif.); sequencing of DNA nanoballs (Complete Genomics, Inc., Mountain View, Calif.);
  • T m is used in reference to the “melting temperature.”
  • the melting temperature is the temperature at which a population of double-stranded nucleic acid molecules becomes half dissociated into single strands.
  • nucleic acid refers to one or more nucleic acids
  • assay includes reference to equivalent steps and methods known to those skilled in the art, and so forth.
  • the assay systems of the invention provide spatially-encoded, multiplexed assays comprising 1) an assay capable of high levels of multiplexing with an efficient spatial encoding scheme; 2) instrumentation capable of delivering reagents according to a spatial pattern; and 3) decoding determined by a readout that is digital in nature.
  • the assay systems of the invention detect the presence or absence and relative amount of a biological target or biological activity indicative of a biological target, as well as the location of the biological target or activity in a biological sample, e.g., a tissue section or other biological structure disposed upon a support such as a microscope slide or culture dish.
  • the assay system further provides instrumentation with an ability to deliver reagents in a spatially-defined pattern.
  • This instrumentation together “with software, reagents and protocols, provides a key component of the highly innovative assay system of the invention, allowing for measurement of numerous biological targets or activities in a meaningful spatial environment, including gene expression and peptide localization,
  • An encoding scheme used in these assay systems allows one to determine the location of biological targets or activity (or lack thereof) in the biological samples after the products of the multiplexed assay are removed from the biological sample and pooled for analysis. Decoding of the encoding scheme can be performed by, e.g., next-generation sequencing, which easily provides millions to trillions of data points at low cost.
  • the assay results such as the amount or activity of biological targets can then be mapped back to specific location in the biological sample.
  • the assay systems open a new analytical window into the complex spatial patterns of cellular function and regulation in biological samples.
  • a biological sample affixed to a support contains biological targets of interest.
  • biological targets can include any molecule of interest, such as nucleic acids (including, e.g, RNA transcripts, genomic DNA sequences, cDNAs, amplicons, or other nucleic acid sequences) and proteins, enzymes and the like.
  • encoded probes are delivered to the biological sample according to a known spatial pattern. Encoded probes comprise probes, which can interact “with biological targets of interest, and coding tags, which identify the positions in the sample of the biological targets being assayed, and thus can be used to link assay results back to locations in the sample. Coding tags in most embodiments are oligonucleotides. However, coding tags may also be mass tags, fluorescent labels, or other moieties.
  • the probe and coding tag portions of the encoded probe are pre-coupled before being delivered to the biological sample.
  • both the probe and coding tag sequence can be synthesized as a single oligonucleotide.
  • the probe and coding tag portions of the encoding probes can be synthesized or obtained separately and combined before delivery to the biological sample (e.g., two separate oligonucleotides can be synthesized and coupled by, e.g., ligation; or an antibody and an oligonucleotide can be prepared separately and conjugated before delivery to the biological sample). Also, as is described in FIGS.
  • the probes and the coding tags are synthesized separately, and are delivered to the biological sample at different steps (e.g., probes first and coding tags thereafter, or vice versa) in the assay.
  • the encoded probes are allowed to react or interact with the biological targets, i.e., conditions are provided to allow e.g., oligonucleotides to hybridize to nucleic acid targets, enzymes to catalyze reactions with protein targets, antibodies to bind epitopes, etc.
  • the encoded probes are typically oligonucleotides and hybridize to the target nucleic acids.
  • the encoded probes typically are aptamers, small molecules, or oligonucleotide-conjugated proteins that interact with target proteins by binding to them or by reacting with them (that is, one of the proteins is a substrate for the other).
  • Encoding oligonucleotides may be coupled to the probes (proteins) by conjugation, chemical or photo-crosslinking via suitable groups and the like.
  • the encoded probes that interacted with the biological targets must be separated from the encoded probes that did not interact with the biological targets at step 140.
  • the separation can be accomplished by, e.g., washing the unhybridized encoded probes from the sample.
  • washing steps can be used to remove low affinity binders.
  • the probe is transformed via interaction with the target, e.g., in the case of a peptide, e.g., via cleavage by a protease or phosphorylation by a kinase
  • an antibody or other affinity capture agent can be used to capture probes that were transformed by addition of a moiety (e.g., a phosphate group).
  • the transformed probes can be separated, e.g., by capturing the non-transformed probes via a tag that is removed from the transformed probes during the transformation (e.g., by cleavage), or by adding a new tag at the site of cleavage.
  • the sequence of the reacted and/or interacted encoded probes is determined at step 150 by, preferably, sequencing.
  • the sequence of the encoded probes allows the mapping of the assay results at step 160 back to locations in the biological sample.
  • FIG. 2 provides a simplified overview of an assay system 200 of the present invention embodying an efficient implementation of a combinatorial coding scheme for the encoding of spatial information.
  • the probes are oligonucleotides, but as explained elsewhere, other types of probes can also be used.
  • a biological sample affixed to a support e.g., a tissue sample or other biological structure.
  • one or more oligonucleotide probes are delivered to the biological sample, where the oligonucleotide probes are capable of hybridizing with biological targets in the biological sample.
  • the oligonucleotide probes are allowed to interact with (hybridize to) the nucleic acid targets; that is, appropriate conditions are provided where oligonucleotide probes can hybridize to the target nucleic acids.
  • step 240 the oligonucleotide probes that did not hybridize to target nucleic acids are removed, and thereby separated from oligonucleotide probes that did hybridize to target nucleic acids.
  • separation can be accomplished by, e.g., washing the sample to remove unhybridized oligonucleotide probes.
  • step 250 encoding oligonucleotides (the encoding agents) are delivered to the biological sample according to a chosen spatial pattern, where the encoding oligonucleotides comprise coding tags that are used to encode the location of biological targets in the biological sample. Note that in contrast to the assay system of FIG.
  • the probes and encoding agents are delivered in separate steps.
  • the encoding oligonucleotides are coupled to the oligonucleotide probes to create encoded probes.
  • the probes are oligonucleotides
  • the encoding oligonucleotides may be coupled to the oligonucleotides probes by, e.g., ligation.
  • the information in the encoding oligonucleotides can be transferred by using a DNA polymerase to extend a probe oligonucleotide that acts as a primer, and thereby copy and incorporate the sequence of the encoding oligonucleotides.
  • step 270 the sequence of the coding tags in the encoded probes as well as the sequence or a portion of the sequence of the probe itself is determined, and in step 280, the target nucleic acids are mapped back to the biological sample.
  • the abundance of sequences reveals the relative quantity of biological targets at the location.
  • steps 220 and 250 can be combined, so that a mixture of the probes and encoding oligonucleotides is delivered according to a chosen spatial pattern.
  • Coupling step 260 can then be carried out immediately after the combined steps 220 and 250, or concomitantly with them.
  • step 240 would then occur after step 260.
  • the two key results of this series of steps i.e., the location-specific encoding of probe molecules and the separation of probe molecules based on their ability to interact with corresponding target molecules, can be accomplished with some flexibility in the implementation of the particular steps.
  • the assays of the invention are particularly amenable to combinatorial methods.
  • the present invention provides an ability to look at many different biological targets in many locations, providing the resolution of in situ hybridization with the highly-parallel data analysis of sequencing.
  • the sum of the multiple biological targets being assayed and the multiple sites in the biological sample is greater than 20, in other embodiments, the sum of the multiple biological targets being assayed and the multiple sites in the biological sample is greater than 50, in other embodiments, the sum of the multiple biological targets being assayed and the multiple sites in the biological sample is greater than 100, greater than 500, 1,000, 10,000, 25,000, 100,000, 500,000, 1,000,000. It will be appreciated that, due to the spatial encoding dimension of the invention, even much larger numbers can be contemplated.
  • assaying 10,000 targets per location ⁇ 10,000 locations would generate 1O ,8 different assays, and even larger numbers than these can easily be contemplated, particularly if spatial locations with resolution on the order of that of single cells are utilized.
  • sequences of at least 1,000 encoding probes are typically determined in parallel. More typically, using a digital readout, it is desirable to obtain multiple sequence reads for each assay (defined by a probe and a spatial location code). It is desirable to obtain an average of at least 3 copies per assay, and more typically at least 10 or at least 30 copies per assay, depending on the design of the experiment and requirements of the assay.
  • the sequence of at least 10,000 encoding probes are determined in parallel, or the sequence of at least 100,000, 500,000, 1,000,000, 10,000,000, 100,000,000, 1,000,000,000 or more encoding probes are determined in parallel.
  • the assay portion of the assay systems of the present invention comprise the following general steps: delivering probes and encoding agents where the encoding agents (in some embodiments pre-coupled to the probes) are delivered to the sample according to a known spatial pattern, allowing the probes to interact or react with biological targets in the sample, and, if the probes and encoding agents have not been pre- coupled, coupling the encoding agents to probes.
  • the samples of the present invention include virtually any biological sample or samples that can be affixed to a support or provided essentially in a two-dimensional manner, where the ability to tie an assayed biological target or activity back to the location within the biological sample is important.
  • exemplary biological samples include tissue sections (e.g., including whole animal sectioning and tissue biopsies), cell populations on slides or culture dishes, and the like.
  • the assay systems of the invention are particularly advantageous in that they are compatible with numerous biological sample types, including fresh samples, such as primary tissue sections, and preserved samples including but not limited to frozen samples and paraformalin-fixed, paraffin-embedded (FFPE) samples.
  • FFPE paraffin-embedded
  • An important aspect of the assay systems of the invention is that the biological samples are immobilized on a substrate surface having discrete, independently measurable areas.
  • the biological targets to be detected can be any biological molecules including but not limited to proteins, nucleic acids, lipids, carbohydrates, ions, or multicomponent complexes containing any of the above.
  • subcellular targets include organelles, e.g., mitochondria, Golgi apparatus, endoplasmic reticulum, chloroplasts, endocytic vesicles, exocytic vesicles, vacuoles, lysosomes, etc.
  • the assay system is used to analyze nucleic acids, e.g., by genotyping, quantitation of DNA copy number or RNA transcripts, localization of particular transcripts within samples, and the like.
  • FIG. 3 illustrates an overall scheme for an exemplary assay for, e.g., detecting single nucleotide polymorphisms (SNPs) that can be used with the assay system of the invention.
  • SNPs single nucleotide polymorphisms
  • two oligonucleotide probes are provided.
  • Each oligonucleotide probe comprises a target-specific region (located on either side of the SNP to be analyzed) seen at 305 and 307, and ligation regions, seen at 301 and 303,
  • the oligonucleotide probes are allowed to hybridize to a target nucleic acid (not shown) in the biological sample.
  • one of the oligonucleotide probes is extended to incorporate the SNP sequence and ligated to the other probe to form an extended probe comprising target nucleic acid region 309 and ligation regions 301 and 303.
  • Two encoding agents both comprising a coding tag (seen at 315 and 317), a ligation region (seen at 311 and 313), and a primer region (seen at 319 and 321) are combined with and ligated to the extended probe at step 304 to form an encoded target-specific oligonucleotide.
  • the probes and encoding agents are delivered at separate steps. Doing so allows use of the combinatorial embodiments described infra.
  • the encoding oligonucleotides within a pair of encoding oligonucleotides ligate specifically to one side of the target sequence or the other (i.e., 5′ or 3′ of the target sequence) in step 306.
  • the ligation and primer regions of the encoding oligonucleotides and probes are universal; that is, the set of ligation and primer regions used in constructing the probes and encoding oligonucleotides are constant, and only the target-specific regions of the probes and the coding tags of the encoding oligonucleotides differ.
  • the ligation and primer regions are not universal and differ between probes and encoding agents.
  • each sequencing adapter comprises primer region 319 or 321, compatible with the primer regions 319 and 321 on the encoded probes.
  • first adapter 327 first primer region 319, first coding tag 315, ligation regions 311 and 301, target region 309, ligation regions 313 and 303, second coding tag 317, second primer region 325 and second adapter 329 is now ready for input into a digital high-throughput sequencing process.
  • a combination of extension and ligation reactions are exemplified in FIG. 3 , but it should be appreciated that a variety of reactions may be used to couple the encoding oligonucleotides to the target-specific oligonucleotides, including ligation only (e.g., for oligonucleotides that hybridize to contiguous portions of the target nucleic acid sequence).
  • an assay utilizing an additional oligonucleotide such as in the GOLDENGATE® assay (see Fan, et al., Cold Spring Symp. Quant. Biol., 68:69-78 (2003); (Ilumina, Inc., San Diego, Calif.)), may be employed.
  • the assay system of the invention also can be used to analyze peptides or proteins, the presence of antibodies, enzymatic and other protein activities, posttranslational modifications, active and non-active forms of peptides, as well as peptide isoforms in a biological sample.
  • the probes may comprise an active region of an enzyme, a binding domain of an immunoglobulin, defined domains of proteins, whole proteins, synthetic peptides, peptides with introduced mutations, aptamers and the like.
  • the probes are substrates for enzymes or proenzymes, e.g., kinases, phosphatases, zymogens, proteases, or fragments thereof.
  • the probes are phosphorylation substrates used to detect proteins involved in one or more signal transduction pathways, e.g., a kinase or a phosphatase.
  • the probes are specific protease substrates that associate only with individual proteases or classes of proteases.
  • the probes are different processed forms, isoforms and/or domains of an enzyme.
  • Protein-based probes are typically conjugated or otherwise linked to oligonucleotide encoding agents.
  • the oligonucleotide encoding agents in this case would also include a nucleotide sequence component that allows for identification of the protein probe.
  • the present invention provides assays for evaluating differences in the amount and/or activity of biological targets between different locations in a sample and/or between samples.
  • the method includes determining a plurality of encoded results from the biological sample and evaluating the differences in quantity of the biological targets at each location in the biological sample.
  • a combinatorial approach using pairs of coding tags in the encoding oligonucleotides can be used.
  • the number of oligonucleotides required is dramatically reduced, with a concomitant decrease in cost.
  • FIG. 4 illustrates a general mechanism for one embodiment of a combinatorial encoding scheme of the assay systems of the invention, where nucleic acids in a representative tissue section (shown at 416) are assayed.
  • FIG. 4 at A shows two target-specific/encoding oligonucleotide constructs 420 and 422 (e.g., formed between steps 302 and 304 of FIG. 3 ) specifically bound to a target nucleic acid 402 of interest.
  • the first encoded probe 420 comprises coding tag 408, associated with, e.g., a universal priming site for amplification of the assay products or an adapter to enable identification of the coding identifiers using sequencing technologies 404.
  • the second encoded probe 422 comprises coding tag 406, associated with, e.g., a universal priming site for amplification of the assay products or an adapter to enable identification of the coding identifiers using sequencing technologies 410.
  • FIG. 4 at B shows the spatial pattern that may be used for twenty different coding tags, a1 through a10 (coding tag 406 on encoded probe 420) and b1 through b10 (coding tag 408 encoded probe 422).
  • Coding tag a1. is deposited on the biological sample in ten discrete areas or spots (shown as the first horizontal line of spots in 412).
  • Coding tag a2 is deposited on the biological sample in ten spots on the second horizontal line in 412.
  • Coding tag a3 is deposited on the biological sample in ten spots on the third horizontal line in 412, and so on. Whereas the “a” tags are deposited in ten horizontal rows, the “b” tags are deposited in ten vertical rows as shown in 414.
  • FIG. 4 at C shows a representative tissue section 416 coincident with coding tag grid 418.
  • the arrows show how the “a” coding tags and the “b” coding tags are deposited on grid 418 that is coincident with tissue section 416. If, once sequenced, coding tags a1 and b4, e.g., are associated with a target nucleic acid sequence, then that target nucleic acid sequence (i.e., biological target) was present in the tissue section at location a1, b4.
  • target nucleic acid sequence i.e., biological target
  • FIG. 5 provides a simplified, specific example of the encoding scheme of the assay systems of the invention.
  • FIG. 5 shows encoding oligonucleotides 510, comprising a1, a2, a3, a4 and b1, b3, b3 and b4.
  • Target-specific oligonucleotides (TSOs) probes 1 and 2 are shown at 520 .
  • a deposit or dispensing scheme is shown at 530 .
  • TSOs target-specific oligonucleotides
  • encoding oligonucleotides a1 through a4 are deposited in spots in a pattern (here, in a vertical pattern), and encoding oligonucleotides b1 through b4 are deposited in spots in a pattern (here, a horizontal pattern).
  • the grid though shown as a square with spots is actually a deposition pattern on a biological sample (not shown) such as tissue section 416 shown in FIG. 4 .
  • the target-specific oligonucleotides are delivered to the biological sample, where the target-specific oligonucleotides hybridize to target nucleic acids in the biological sample if target nucleic acids are present. Unhybridized target-specific oligonucleotides are then removed, e.g., by washing. The encoding oligonucleotides are then delivered to the biological sample according to the spatial pattern shown at 530.
  • the encoding oligonucleotides are ligated (or, e.g., extended and ligated) to any target-specific oligonucleotides that hybridized to the target nucleic acid in the biological sample, the ligated constructs are then eluted from the biological sample, pooled, and sequencing adapters are added through, e.g., PCR or ligation, if the sequences were not previously included in the encoding oligonucleotides.
  • the ligated constructs are sequenced by, e.g., high throughput or “next generation” sequencing.
  • a sequence readout was obtained for target-specific oligonucleotide 1 only at a4b1, a4b2, a1b3, a2b3, a3b3, a4b3 and a4b4 (positions shown with horizontal lines).
  • a sequence readout was obtained for target-specific oligonucleotide 2 only at a1b1 (position shown with vertical lines).
  • a sequence readout was obtained for both target-specific oligonucleotides 1 and 2 at positions a2b1, a3b1, a1b2, a2b2, and a3b2 (positions shown with cross-hatching).
  • information relating to relative abundance of the encoded tags can be obtained. For example, if it is found that there are ten times as many a4T1b1 sequences occurring in the data set as compared to a4T1b2 sequences, this would indicate that target nucleic acid sequence 1 is ten times more abundant at the a4T1b1 location than at the a4T1b2 location.
  • nucleotide analysis as shown in FIG. 3 , by ligating the coding tags directly to target-specific oligonucleotides, only 2n target-specific oligonucleotides are needed for n targets.
  • assaying 100 different targets at 10,000 spatial locations would require 2 ⁇ 100 target-specific oligonucleotides and 2 ⁇ 100 encoding oligonucleotides.
  • the total count of assay oligonucleotides would be only 400 (200 target-specific and 200 encoding), not counting universal primers.
  • coding oligonucleotides were not decoupled from the target-specific oligonucleotides, (n ⁇ X positional codes)+(n ⁇ Y positional codes) would be needed, or in the above example, 20,000 oligonucleotides, not counting universal primer sequences.
  • FIGS. 2-5 depict a combinatorial scheme using two encoding agents (coding tags), three, four or more encoding agents and coding tags may be used, and attached to the probe or one another by varying means and in varying combinations of steps.
  • a large amount of information can be generated with even a modest number of assays. For example, five or more biological targets assayed at five or more positions in the sample generates 25 or more combinations.
  • the optimum number of sequence reads per combination depends on the sensitivity and dynamic range required, and can be adjusted, For example, if for each combination on average 100 reads are sampled, the total for 25 combination is 25,000 reads. If 1,000 targets are assayed at 1,000 locations with an average sampling depth of 1,000, then 10 9 reads are required.
  • the assay areas may be physically separated using barriers or channels.
  • Pumps generally include any mechanism for moving fluid and/or reagents disposed in fluid.
  • the pump can be configured to move fluid and/or reagents through passages with small volumes (i.e., microfluidic structures).
  • the pump can operate mechanically by exerting a positive or negative pressure on fluid and/or on a structure carrying fluid, electrically by appropriate application of an electric field(s), or both, among other means.
  • Exemplary mechanical pumps may include syringe pumps, peristaltic pumps, rotary pumps, pressurized gas, pipettors, etc.
  • Mechanical pumps may be micromachined, molded, etc.
  • Exemplary electrical pumps may include electrodes and may operate by electrophoresis, electroendoosmosis, electrocapillarity, dielectrophoresis (including traveling wave forms thereof), and/or the like.
  • Valves generally include any mechanism for regulating the passage of fluid through a channel.
  • Valves can include, for example, deformable members that can be selectively deformed to partially or completely close a channel, a movable projection that can be selectively extended into a channel to partially or completely block a channel, an electrocapillary structure, and/or the like.
  • An open gasket can be attached to the top of the biological sample and the sample and reagents can be injected into the gasket.
  • Suitable gasket materials include, but are not limited to, neoprene, nitrile, and silicone rubber.
  • a watertight reaction chamber may be formed by a gasket sandwiched between the biological sample on the substrate and a chemically inert, water resistant material such as, but not limited to, black-anodized aluminum, thermoplastics (e.g., polystyrene, polycarbonate, etc), glass, etc.
  • the assay system comprises imaging means to determine features and organization of the biological sample of interest.
  • the images obtained may be used to design the deposition pattern of the reagents.
  • Imaging means are optional, as an individual can instead view the biological sample using, e.g., a microscope, analyze the organization of the biological sample, and specify a spatial pattern for delivery assay reagents.
  • the delivery system can comprise a microcircuit arrangement including an imager, such as a CCD or IGFET-based (e.g., CMOS-based) imager and an ultrasonic sprayer for reagent delivery such as described in US Pub. No. 20090197326, which is incorporated herein by reference.
  • FIGS. 4 and 5 illustrate using a x,y grid configuration, other configurations can be used, such as, e.g., following the topology of a tissue sample; targeting certain groups of cells, cell layers and/or cell types in a tissue, and the like.
  • the reagent delivery system controls the delivery of reagents to specific patterns on a biological sample surface using semiconductor techniques such as masking and spraying. Specific areas of a biological sample can be protected from exposure to reagents through use of a mask to protect specific areas from exposure.
  • the reagents may be introduced to the biological sample using conventional techniques such as spraying or fluid flow. The use of masked delivery results in a patterned delivery scheme on the substrate surface.
  • the reagent delivery instrumentation is based on inkjet printing technology.
  • inkjet printing technology There are a variety of different ink-jetting mechanisms (e.g., thermal, piezoelectric) and compatibility has been shown with aqueous and organic ink formulations.
  • Sets of independently actuated nozzles can be used to deliver multiple reagents at the same time, and very high resolutions are be achieved.
  • an informative image of the biological sample to be assayed may be used to assist in the reagent delivery methods and associated encoding scheme.
  • Sample regions of the biological sample can be identified using image processing (e.g., images of cell types differentiated by immunohistochemistry or other staining chemistries) integrated with other features of the assay system.
  • software is used to automatically translate image information into a reagent delivery pattern.
  • a mechanism to register and align very precisely the biological sample for reagent delivery is thus an important component of the assay systems of the invention. Mechanisms such as the use of fiducial markers on slides and/or other very accurate physical positioning systems can be adapted to this purpose.
  • the invention preferably comprises a complete suite of software tailored to the assay system.
  • oligonucleotide design software is used to design the encoding nucleotides (and in embodiments where nucleic acids are assayed, the target-specific oligonucleotides) for the specific assay to be run, and may be integrated as a part of the system.
  • algorithms and software for reagent delivery and data analysis i.e., sequence analysis
  • sequence analysis may be integrated to determine assay results. Integrated data analysis is particularly useful, as the type of dataset that is generated may be massive as a consequence of scale.
  • Algorithms and software tools that are specifically designed for analysis of the spatially-associated data generated by the assay systems, including pattern-analysis software and visualization tools, enhance the value of the data generated by the assay systems.
  • the assay system comprises processes for making and carrying out the quality control of reagents, e.g., the integrity and sequence fidelity of oligonucleotide pools.
  • reagents are formulated according to factors such as volatility, stability at key temperatures, and chemical compatibility for compatibility with the reagent delivery instrumentation and may be analyzed by instrumentation integrated within the assay system.
  • the encoded probes can be amplified and hybridized to a microarray. This would require separate amplification reactions to be carried out, in which each amplification is specific to a particular spatial code or subset of codes, accomplished by using code-specific primers. Each amplification would also incorporate a different resolvable label (e.g. fluorophor). Following hybridization, the relative amounts of a particular target mapping to different spatial locations in the sample can he determined by the relative abundances of the resolvable labels.
  • a resolvable label e.g. fluorophor
  • the resulting coding tags according to the assay system are substrates for high-throughput, next-generation sequencing, and highly parallel next-generation sequencing methods are used to confirm the sequence of the coding tags, for example, with SOLiDTM technology (Life Technologies, Inc.) or Genome Ananlyzer (Illumina, Inc.).
  • next-generation sequencing methods can be carried out, for example, using a one pass sequencing method or using paired-end sequencing.
  • Next generation sequencing methods include, but are not limited to, hybridization-based methods, such as disclosed in e.g., Drmanac, U.S. Pat. Nos. 6,864,052; 6,309,824; and 6,401,267; and Drmanac et al, U.S.
  • 3-dimensional patterns of gene expression are determined by analyzing a series of tissue sections, in a manner analogous to image reconstruction in CT scanning,
  • a method can be used to measure changes in gene expression in disease pathology, e.g., in cancerous tissue and/or a tissue upon injury, inflammation or infection.
  • disease pathology e.g., in cancerous tissue and/or a tissue upon injury, inflammation or infection.
  • assay systems of the invention more detailed information on gene expression and protein localization in complex tissues is obtained, leading to new insights into the function and regulation both in normal and diseased states, and provides new hypotheses that can be tested.
  • the assay systems also provide a novel approach to analysis of somatic variation, e.g., somatic mutations in cancer or variability in response to infectious organisms.
  • somatic variation e.g., somatic mutations in cancer or variability in response to infectious organisms.
  • tumors are typically highly heterogeneous, containing cancer cells as well as genetically normal cells in an abnormal local environment. Cancer cells undergo mutation and selection, and in this process it is not unusual for local clones to develop. Identifying relatively rare somatic mutations in the context of tumors may enable the study of the role of key mutations in the selection of clonal variants.
  • Transcriptional patterns associated with angiogenesis, inflammation, or other cancer-related processes in both cancer and genetically normal cells can be analyzed for insights into cancer biology and assist in the development of new therapeutic agents for the treatment of cancers.
  • individuals have varying susceptibility to infectious organisms, and the assay systems of the invention can be used to study the interaction between microbes and tissues or the various cell types within the tissue.
  • This encoding scheme uses both molecule-specific binding agents and coding identifiers to provide a practical and cost-effective determination of information on multiple biological molecules, including specific positional information of such molecules in a biological sample, e.g., a tissue section.
  • the single molecule detection analysis using the encoding system also allows relative amounts of biological molecules to be detected, thus providing information on expression levels, sequestering in specific locales, and the like.
  • the assay system and methods of the invention are based on relational, solid-state substrates with positions that represent specific spatial locations within a biological sample, e.g., a cell, organelle or tissue.
  • a biological sample e.g., a cell, organelle or tissue.
  • the ability to use encoding features to represent locations allows high-throughput analysis of the presence or absence, and relative amount, of a biological molecule at more than one spatial location in a sample.
  • the encoding features also allow provide assaying of multiple biological molecules at these multiple locations simultaneously.
  • a primary feature of the invention is the preservation of the spatial organization of elements in a sample of interest through the use of an encoding scheme.
  • the assay may be designed to preserve the relative position of cells in a tissue, and the assay may interrogate the individual cells for genomic DNA variation (including epigenetic modifications), and RNA and protein expression.
  • the binding agents are immobilized directly to the substrate surface, and the location of the binding agents is known or determined prior to use of the substrate surface in the assay system.
  • the binding agents are immobilized onto beads or other separate structural elements that are then provided in known locations on the substrate surface.
  • the binding agents may be provided in or on features of the substrate surface, e.g., provided in wells or channels.
  • the binding agents are nucleic acids immobilized directly or indirectly to the substrate surface, e.g., directly through the use of amino groups on the substrate surface or indirectly through the use of a linker.
  • the location of the nucleic acid sequences is known or determined prior to use of the substrate surface in the assay system.
  • the nucleic acids may be immobilized directly or indirectly onto beads that are then provided in known locations on the substrate surface.
  • the nucleic acids may be provided in or on features of the substrate surface, e.g., provided in wells.
  • nucleic acid sequencing any methods of sequence determination can be used, e.g., sequencing, hybridization and the like.
  • nucleic acid sequencing and preferably next-generation sequencing, is used to decode the spatial encoding scheme in the assay system of the invention. This provides a very wide dynamic range for very large numbers of assays, allowing for efficient multiplexing.
  • the assay utilizes two or more oligonucleotides, the oligonucleotides comprising a universal primer region and a region that correlates specifically to a single spatial pattern within the spatial encoding scheme.
  • the assay comprises two allele specific oligonucleotides and one locus specific oligonucleotides. These oligonucleotides allow the identification of specific SNPs, indels or mutations within an allele. This is useful in the identification of genetic changes in somatic cells, genotyping of tissues, and the like.
  • the biological molecules to be detected can be any biological molecules such as proteins, nucleic acids, lipids, carbohydrates, ions, or multicomponent complexes containing any of the above.
  • Further examples of subcellular objects include organelles, e.g., mitochondria, Golgi apparatus, endoplasmic reticulum, chloroplast, endocytic vesicle, exocytic vesicles, vacuole, lysosome, etc.
  • the assay systems of the invention are particularly advantageous in that they are compatible with numerous samples types, such as fresh samples, such as primary tissue sections, and preserved samples including but not limited to frozen samples and paraformalin-fixed, paraffin-embedded (FFPE) samples.
  • An important aspect of the assay systems of the invention is that the binding agents are immobilized on a substrate surface in discrete, independently measureable areas. These discrete areas can be formed by spatially selective deposition of the binding agents on the substrate surface. Numerous methods can be used for the deposition of the agent and the coding identifiers associates with the agent. For example, the coding identifiers can be delivered together or separately from the agent.
  • Examples of methods that can be used for deposition of agents and/or coding identifiers onto the substrate surface include, but are not limited to, ink jet spotting, mechanical spotting by means of pin, pen or capillary, micro contact printing, fluidically contacting the measurement areas with the biological or biochemical or synthetic recognition elements upon their supply in parallel or crossed micro channels, upon exposure to pressure differences or to electric or electromagnetic potentials, and photochemical or photolithographic immobilization methods.
  • the substrates may be arranged into segments of one or more measurement areas for reagent distribution and agent determination. These regions may be physically separated using barriers or channels. They may still comprise several additional discrete measurement areas with agents that are different or in different combination from each other.
  • the present invention provides a method, e.g., a machine-based method, for evaluating changes in the presence and/or location of a biological molecule over time.
  • the method includes providing a plurality of encoded array results representative of the biological molecule over time and evaluating the differences in detection and/or localization of the biological molecules.
  • the detection of nucleic acids uses two allele-specific oligonucleotides and a locus specific oligonucleotide.
  • the assay methods are carried out according to the strategy outlined in FIG. 2 using next-generation sequencing or another highly parallel nucleic acid assay technology.
  • a set of two oligonucleotides is designed to hybridize to each target sequence, with a common oligonucleotide and two unique coding identifiers.
  • the allele can be determined, e,g, by primer extension of the locus specific oligonucleotide.
  • an amplifiable template is formed with universal primer sequences at either end.
  • Assay oligonucleotides are annealed to a template and enzymatic reactions are used to join the two oligonucleotides only when both are correctly annealed.
  • the detection techniques and read out parameters used in this system of the invention include a much shorter tag than the oligonucleotides used in the assays that are based on capture by hybridization. These shorter tags are designed to be read out by sequencing or, preferably, used to ligate codes onto both ends of the fragment as illustrated in FIG. 2 .
  • two target-specific assay oligonucleotides are ligated together 202 following in situ hybridization to target sequences.
  • encoding oligonucleotides containing tag sequence sets X and Y are ligated 204 to the target specific oligonucleotides.
  • Oligonucleotides containing X ligate specifically to one side of the targeting construct and oligonucleotides containing Y ligate to the other.
  • the oligonucleotides contain universal primer sites P1 and P2.
  • the constructs are eluted and, optionally, sequencing adapters can be attached 206, e.g., via PCR.
  • the final construct created from the assay method is a substrate for next-generation sequencing, and highly parallel next-generation sequencing methods are used to confirm the sequence of constructs.
  • sequencing methods can be carried out, for example, using a one pass sequencing method or using paired-end sequencing.
  • Next generation sequencing methods include, but are not limited to, hybridization-based methods, such as disclosed in Drmanac, U.S. Pat. Nos. 6,864,052; 6,309,824; and 6,401,267; and Drmanac et al, U.S. patent publication 2005/0191656, and sequencing by synthesis methods, e.g., Nyren et al, U.S. Pat. No. 6,210,891; Ronaghi, U.S. Pat. No.
  • a combinatorial approach using pairs of oligonucleotides can be used. For example, with only two sets of 100 codes, a substrate can theoretically encode up to 10,000 locations. The number of assay oligonucleotides required is dramatically reduced, the cost decreased, and the robustness of the approach increased by decoupling the coding sequences from the genome-specific sequences.
  • Alternative assay formats can also be used (e.g. ligation or primer extension followed by ligation).
  • a large amount of information can be obtained even using five or more positions interrogated for five or more biological molecules.
  • large amounts of information can be obtained, both in terms of locations and/or specific biological
  • multiple locations are interrogated for two or more biological molecules.
  • ⁇ 1,000 reads may be sampled, for a total of ⁇ 10E9 reads for 10E6 datapoints.
  • the assay system of the invention can be used to analyze biological molecules using peptide agents that are associated with the substrate surface in a spatial pattern.
  • peptide agents may comprise an active region of an enzyme, a binding domain of an immunoglobulin, defined domains of proteins, whole proteins, synthetic peptides, peptides with introduced mutations, etc.
  • the assay system of the invention allows the identification and spatial location of various forms of peptides, including isoforms and peptides that have undergone posttranslational modification. Importantly, certain aspects of the invention allow the identification of active versus non-active forms of such peptides in a sample. This allows the identification of the presence or absence of specific peptide isoforms, and also acts as a proxy for identification of peptide activity in a sample.
  • the binding agents associated with the substrate surfaces of the assay system include substrates for enzymes or proenzymes, e.g., a kinase, a phosphatase, a zymogen, a protease, or a fragment thereof.
  • the binding agents associated with the substrate surfaces are phosphorylation substrates used to detect proteins involved with one or more signal transduction pathways, e.g., a kinase or a phosphatase.
  • the binding agents are specific protease substrates that associate only with individual or classes of proteases.
  • the binding agents on the substrate surface are different processed forms, isoforms and/or domains of an enzyme.
  • the reagent delivery system of the invention can be any system that allows the delivery of reagents to discrete portions of the array in order to keep the integrity of the defined spatial patterns of the encoding scheme. Such discrete delivery can be achieved in a number of different ways.
  • US Appln Nos. 20070166725 and 20050239192 disclose certain general-purpose fluidics tools that can be used with the assay systems of the invention. These allow the precise manipulation of gases, liquids and solids to accomplish very complex analytical manipulations with relatively simple hardware.
  • one or more flow-cells can be attached to the substrate from above.
  • the flow-cell can include inlet and outlet tubes connected thereto and optionally an external pump can be used to deliver the sample or reagents to the flow-cell and across the substrate.
  • the flow cell is configured to deliver reagents only to certain portions of the array, restricting the amount and type of reagent delivered to any specific section of the array.
  • a microfluidic system can be integrated into the substrate or externally attached on top of the substrate.
  • Microfluidic passages for holding and carrying fluid can be formed on and/or above the planar substrate by a fluidics layer abutted to the substrate.
  • Fluid reagents can be selected according to selective opening and closing of valves disposed between reagent reservoirs.
  • Pumps generally include any mechanism for moving fluid and/or reagents disposed in fluid.
  • the pump can be configured to move fluid and/or reagents through passages with small volumes (i.e., microfluidic structures).
  • the pump can operate mechanically by exerting a positive or negative pressure on fluid and/or on a structure carrying fluid, electrically by appropriate application of an electric field(s), or both, among others.
  • Exemplary mechanical pumps may include syringe pumps, peristaltic pumps, rotary pumps, pressurized gas, pipettors, etc.
  • the mechanical pumps may be micromachined, molded, etc.
  • Exemplary electrical pumps can include electrodes and may operate by electrophoresis, electroendoosmosis, electrocapillarity, dielectrophoresis (including traveling wave forms thereof), and/or the like.
  • Valves generally include any mechanism for regulating the passage of fluid through a channel.
  • the valves can include, for example, deformable members that can be selectively deformed to partially or completely close a channel, a movable projection that can be selectively extended into the channel to partially or completely block the channel, an electrocapillary structure, and/or the like.
  • an open gasket can be attached to the top of the substrate and the sample and reagents can be injected into the gasket.
  • gasket materials include, but are not limited to, neoprene, nitrile, and silicone rubber.
  • a watertight reaction chamber formed by a gasket sandwiched between the substrate and a chemically inert, water resistant material such as, but not limited to, black-anodized aluminum, thermoplastics (e.g., polystyrene, polycarbonate, etc), glass, etc.
  • the delivery system can comprise a microcircuit arrangement including an imager, such as a CCD or IGFET-based (e.g., CMOS-based) imager and an ultrasonic sprayer for reagent delivery such as described in US Appln No. 20090197326, which is incorporated herein by reference.
  • an imager such as a CCD or IGFET-based (e.g., CMOS-based) imager and an ultrasonic sprayer for reagent delivery such as described in US Appln No. 20090197326, which is incorporated herein by reference.
  • the reagent delivery system controls the delivery of reagents to specific patterns on a substrate surface using semiconductor techniques such as masking and spraying. Specific areas of a substrate surface can be protected from exposure to reagents through use of a mask to protect specific areas from exposure.
  • the reagents may be introduced to the substrate using conventional techniques such as spraying or fluid flow. The use of the masked substrate delivery results in a patterned delivery scheme on the substrate surface.
  • the reagent delivery instrumentation is based on inkjet printing technology.
  • inkjet printing technology There are a variety of different ink-jetting mechanisms (e.g., thermal, piezoelectric) and compatibility has been shown with aqueous and organic ink formulations.
  • Sets of independently actuated nozzles can be used to deliver multiple reagents at the same time, and very high resolutions can be achieved.
  • an informative image of the biological section to be analyzed can be used to assist in the reagent delivery methods and associated encoding scheme.
  • Sample regions can be identified using image processing (e.g., images of cell types differentiated by immunohistochemistry or other staining chemistries) integrated with the other features of the assay system.
  • software is used to automatically translate this information into a reagent delivery pattern.
  • a mechanism to register and align very precisely the biological sample in a targeting system is thus a preferred component of the assay systems of the invention. Mechanisms such as the use of fiducial markers on slides and other very accurate physical positioning systems can be adapted to this purpose.
  • Additional software components will also be key components that will be part of a complete assay system.
  • the invention thus preferably comprises a complete suite of software tailored to the assay system.
  • oligonucleotide design software will be customized for the specific assay to be run, and may be integrated as a part of the system.
  • algorithms and software for data analysis may be integrated to assist in determination of results of the assays. This can be especially useful, as the type of dataset that will be generated will be novel, particularly as a consequence of scale.
  • the ability to provide algorithms and software tools that are specifically designed for analysis of spatially-associated data for significant patterns, including pattern-analysis software and visualization tools, is a novel feature that will enhance the value of the data generated by the assay systems.
  • the assay system will comprise processes for making and carrying out quality control of reagents, e.g., the integrity and sequence fidelity of oligonucleotide pools.
  • reagents will need to be formulated for compatibility with the reagent delivery instrumentation. Factors such as volatility, stability at key temperatures, and chemical compatibility can be optimized by those skilled in the art upon reading the present disclosure.
  • 3-dimensional patterns of expression can be determined by analyzing a series of tissue sections, in a manner analogous to image reconstruction in CT scanning. This can be used to measure changes in gene expression in disease pathology, e.g., in cancerous tissue and/or a tissue upon injury, inflammation or infection.
  • disease pathology e.g., in cancerous tissue and/or a tissue upon injury, inflammation or infection.
  • assay systems of the invention more detailed information on gene expression and protein localization in complex tissues can be obtained. This may lead to new insights into the function and regulation both in normal and diseased states, and is likely to provide new hypotheses that can be tested.
  • a system of the invention may enable some of the insights gained from many individual studies and larger programs like ENCODE (Birney et al., 2007) and modENCODE to be integrated at the tissue level.
  • ENCODE Billirney et al., 2007
  • modENCODE modENCODE
  • the technology of the invention will allow a great increase in the sensitivity of detecting rare mutations.
  • the reason is that signal to noise can be dramatically increased because the approach of the invention assays a small location in any given reaction.
  • the sample is treated in bulk, i.e. nucleic acids are extracted from many cells into a single pool.
  • nucleic acids are extracted from many cells into a single pool.
  • a mutation is present in 1 cell in 10,000, it must be detected against a background of normal DNA from 10,000 cells.
  • many cells can be analyzed, but individual cells or small groups of cells would be identified by the spatial coding system. Therefore, the background can be reduced by orders of magnitude, greatly increasing sensitivity.
  • the sites comprise different target compositions to show that the assay readout matches the expected composition of each site.
  • the assay readout is then determined to show that the detected regions match the expected signal after spatial decoding.
  • the code space is large enough (2 24 ) so that even a few errors would not result in different codes being mixed up.
  • this design allows identification of errors and allows an estimation not only of accuracy of spatial encoding but also of accuracy calling the presence or absence of target sequences.
  • Genomic DNA is assayed as a proof of concept for assaying RNA, as it provides a way to establish a single-copy reference signal.
  • a working assay is developed for FFPE samples, it is adapted to an RNA assay.
  • assay oligonucleotide concentrations are assayed to ensure compatibility with high multiplexing. Assuming a cell diameter of 10 microns, and delivery of a 10 micron diameter reagent droplet to an individual cell, the volume of the droplet will be ⁇ 500 ⁇ l and can contain ⁇ 3 ⁇ 10 11 molecules at a 1 ⁇ M concentration. To assay 1,000 target sequences in 10,000 cells, 2,000 targeting oligonucleotides would be required in a droplet. Therefore, each droplet could contain ⁇ 160 million copies of each assay oligo, a vast excess over the few thousand target sequences in a cell.
  • Example 4 Adapting the Assay to a Biological Sample
  • a control RNA template is immobilized to a solid support in order to create an artificial system.
  • the assay is performed using T4 DNA ligase, which can repair nicks in DNA/RNA hybrids. Assays are carried out on matched slides, or different sections of the same slide, where in one case gDNA is assayed and in the other RNA is assayed.
  • Assaying gDNA the slide can be pretreated with RNase, and when assaying RNA the slide is pretreated with DNase. Results of the assay are confirmed by extracting gDNA or RNA and quantitating the relative amounts by qPCR or RT-qPCR respectively.
  • RNA assays In order make the tissue section RNA assays as informative as possible, pre-existing information on expression levels in specific tissues to target transcripts across a range of abundances are used in the assay design. Both high abundance transcripts, as well as some medium and low abundance transcripts, are targeted to enable an initial assessment of the quantitative performance characteristics of the assay.

Abstract

The present invention provides assays and assay systems for use in spatially encoded biological assays. The invention provides an assay system comprising an assay capable of high levels of multiplexing where reagents are provided to a biological sample in defined spatial patterns; instrumentation capable of controlled delivery of reagents according to the spatial patterns; and a decoding scheme providing a readout that is digital in nature.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 16/430,015, filed Jun. 3, 2019, which is a continuation of U.S. patent application Ser. No. 16/414,213, filed on May 16, 2019, which is a continuation of U.S. patent application Ser. No. 16/402,098 filed May 2, 2019, which is a continuation of U.S. patent application Ser. No. 16/276,235 filed Feb. 14, 2019, which is a continuation application of U.S. patent application Ser. No. 15/187,661 filed Jun. 20, 2016 (now U.S. Pat. No. 10,308,892), which is a continuation of 13/080,616 filed Apr. 5, 2011 (now U.S. Pat. No. 9,371,598), which claims the benefit of U.S. Provisional Patent Application No. 61/321,124, filed Apr. 5, 2010, each of which is incorporated herein by reference in its entirety.
  • FIELD OF THE INVENTION
  • This invention relates to assays of biological molecules, and more particularly to assays for determining spatial distributions of a large number of biological molecules in a solid sample simultaneously.
  • BACKGROUND OF THE INVENTION
  • In the following discussion certain articles and methods will be described for background and introductory purposes. Nothing contained herein is to be construed as an “admission” of prior art. Applicant expressly reserves the right to demonstrate, where appropriate, that the articles and methods referenced herein do not constitute prior art under the applicable statutory provisions.
  • Comprehensive gene expression analysis and protein analysis have been useful tools in understanding mechanisms of biology. Use of these tools has allowed the identification of genes and proteins involved in development and in various diseases such as cancer and autoimmune disease. Conventional methods such as in situ hybridization and other multiplexed detection of different transcripts have revealed spatial patterns of gene expression and have helped shed light on the molecular basis of development and disease. Other technologies that have enabled the quantitative analysis of many RNA sequences per sample include microarrays (see Shi, et al., Nature Biotechnology, 24(9):1151-61 (2006); and Slonim and Yanai, Plos Computational Biology, 5(10):e1000543 (2009)); serial analysis of gene expression (SAGE) (see Velculescu, et al, Science, 270(5235):484-87 (1995)), high-throughput implementations of qPCR (see Spurgeon, et al., Plos ONE, 3(2):e1662 (2008)) and in situ PCR (see Nuovo, Genome Res., 4:151-67 (1995)). As useful as these methods are, however, they do not enable simultaneous measurement of the expression of many genes or the presence and/or activity of multiple proteins at many spatial locations in a sample. Laser capture microdissection has permitted the analysis of many genes at a small number of locations, but it is very expensive, laborious, and does not scale well. Certain PCR assays in a 2D format preserve spatial information (see Armani, et al., Lab on a Chip, 9(24): 3526-34 (2009)), but these methods have low spatial resolution because they rely on physical transference of tissue into wells, which also prevents random access to tissue samples and high levels of multiplexing.
  • At present, no practical method exists to analyze at high resolution the spatial expression patterns of large numbers of genes, proteins, or other biologically active molecules simultaneously. There is thus a need for reproducible, high-resolution spatial maps of biological molecules in tissues. The present invention addresses this need.
  • SUMMARY OF THE INVENTION
  • This Summary is provided to introduce a selection of concepts in a simplified form that are further described below in the Detailed Description. This Summary is not intended to identify key or essential features of the claimed subject matter, nor is it intended to be used to limit the scope of the claimed subject matter. Other features, details, utilities, and advantages of the claimed subject matter will be apparent from the following written Detailed Description including those aspects illustrated in the accompanying drawings and defined in the appended claims.
  • The invention encompasses assay systems that provide high-resolution spatial maps of biological activity in tissues. The assay system comprises an assay capable of high levels of multiplexing where encoded probes are provided to a biological sample in defined spatial patterns; instrumentation capable of controlled delivery of reagents according to the spatial patterns; and a decoding scheme providing a readout that is digital in nature. In short, the present invention provides the ability to look at many biological targets in many locations, providing the resolution of in situ hybridization with the highly- parallel data analysis of sequencing.
  • Thus, in some embodiments, the invention provides an assay system to determine spatial patterns of abundance or activity or both of multiple biological targets at multiple sites in a sample, where the assay system performs the following steps: providing a sample affixed to a support; delivering encoded probes for the multiple biological targets to the multiple sites in the sample in a known spatial pattern, where each encoded probe comprises a probe region that may interact with the biological targets and a coding tag that identifies a location of the site to which the encoded probe was delivered; allowing the encoded probes to interact with the biological targets; separating encoded probes that interact with the biological targets from encoded probes that do not interact with the biological targets; determining all or a portion of a sequence of the encoded probes, and associating the abundance or activity or both of the multiple biological targets to the locations of the sites in the sample.
  • In particular aspects of the invention the biological targets comprise nucleic acids and the encoded probes are oligonucleotides, and in some aspects, there are two encoded probes for each of the multiple nucleic acid targets. In some aspects, the multiple biological targets comprise proteins, the probe regions of the encoding probes are proteins and the coding tags comprise oligonucleotides. In some aspects the multiple biological targets comprise enzymes. In some aspects the probe regions of the encoded probes comprise antibodies, aptamers or small molecules.
  • Some aspects of the assay system further comprise an amplification step between the separating step and the determining step. In some aspects, the determining step is performed by nucleic acid sequencing, and in preferred aspects, the sequencing is high- throughput digital nucleic acid sequencing.
  • In some aspects of the invention, the product of the multiple biological targets being assayed and the multiple sites in the sample is greater than 20, in some aspects product of the multiple biological targets being assayed and the multiple sites in the sample is greater than 50, in some aspects the product of the multiple biological targets being assayed and the multiple sites in the sample is greater than 75, 100, 150, 500, 750, 1,000, 5,000, 10,000, 25,000, 50,000, 100,000, 500,000, or 1,000,000 or more. In other aspects, the sequence of at least fifty thousand encoding probes are determined in parallel, in other aspects the sequence of at least one hundred thousand encoding probes are determined in parallel, in some aspects the sequence of at least five hundred thousand encoding probes are determined in parallel, and in some aspects the sequence of at least one million, ten million, one hundred million, one billion, ten billion, one hundred billion or more encoding probes are determined in parallel.
  • In some aspects, the known spatial pattern is determined by histological features of the sample. Also in some aspects, software programmed hardware performs at least two steps of the delivering step, the separation step, the determining step and the associating step.
  • In some aspects, the probe regions of the encoded probes are proteins and the separating step is accomplished by encoded probes that interact with the biological targets being captured by an affinity capture agent. In some aspects the probe regions of the encoding probes are nucleic acids and the separating step is accomplished by a washing of the sample.
  • In other embodiments there is provided an assay system to determine spatial patterns of abundance or activity or both of multiple nucleic acid targets at multiple sites in a sample, where the assay system performs the following steps: providing a sample affixed to a support; delivering oligonucleotide probes for multiple nucleic acid targets to the multiple sites in the sample in a known spatial pattern; allowing the oligonucleotide probes to hybridize with the nucleic acid targets; washing unhybridized encoded oligonucleotide probes from the sample; delivering one or more encoding agents to locations of the multiple sites in the sample according to a known spatial pattern, where the combination of encoding agents delivered to each site is different; coupling the encoding agents and the oligonucleotide probes to form encoded probes; determining all or a portion of a sequence of the encoded probes using high-throughput sequencing, and associating the abundance or activity or both of multiple biological targets to the locations of multiple sites in the sample.
  • Other embodiments of the invention provide an assay system to determine spatial patterns of abundance or activity or both of multiple protein targets at multiple sites in a sample, where the assay system performs the following steps: providing a sample affixed to a support; delivering encoded probes for the multiple protein targets to the multiple sites in the sample in a known spatial pattern, where each encoded probe comprises a protein probe region that may interact with the protein targets and a coding tag that identifies a location of the site to which the encoded probe was delivered and the protein probe region of the encoding probe of which the coding tag is part; allowing the encoded probes to interact with the protein targets; separating encoded probes that interact with the protein targets from encoded probes that do not interact with the protein targets; determining all or a portion of a sequence of the encoded probes by high throughput sequencing, and associating the abundance or activity or both of the multiple protein targets to the locations of the multiple sites in the sample.
  • Other embodiments provide an assay system to determine spatial patterns of abundance or activity or both of multiple biological targets at multiple sites in a sample, where the assay system performs the following steps: providing a sample affixed to a support; delivering encoded probes for the multiple biological targets to the multiple sites in the sample in a known spatial pattern, where each encoded probe comprises a probe region that may interact with the biological targets and a coding tag that identifies a location of the site to which the encoded probe was delivered and identifies the biological target; allowing the encoded probes to interact with the biological targets; determining all or a portion of a sequence of the encoded probes, and associating the abundance or activity or both of the multiple biological targets to the locations of the sites in the sample.
  • The assay system of the invention can utilize various detection mechanisms, based on the molecules to be detected and the reagents needed for such detection system. Exemplary methods that can be used with the assay systems of the invention are described in more detail below.
  • DESCRIPTION OF THE FIGURES
  • FIG. 1 provides a simplified overview of the assay system of the present invention.
  • FIG. 2 provides a simplified overview of one embodiment of the assay system of the present invention for detecting nucleic acids.
  • FIG. 3 is a representational depiction of one embodiment of the assay overviewed in FIG. 2.
  • FIG. 4A-C illustrates a general mechanism for one embodiment of a combinatorial encoding scheme of the assay systems of the invention. FIG. 4A shows two target-specific/encoding oligonucleotide constructs specifically bound to a target nucleic acid of interest in a sample. FIG. 4B shows a scheme for delivering twenty different coding tags, a1 through a10 and b1 through b10, to a sample to form a 10×10 coding tag grid. FIG. 4C shows a tissue section sample to which the coding tags are delivered, forming the coding tag grid in the sample.
  • FIG. 5 provides a simplified, specific example of the embodiment of a combinatorial encoding scheme shown in FIG. 4.
  • DEFINITIONS
  • The terms used herein are intended to have the plain and ordinary meaning as understood by those of ordinary skill in the art. The following definitions are intended to aid the reader in understanding the present invention, but are not intended to vary or otherwise limit the meaning of such terms unless specifically indicated.
  • The term “antibody” as used herein is intended to refer to an entire immunoglobulin or antibody or any functional fragment of an immunoglobulin molecule which is capable of specific binding to an antigen (antibodies and antigens are “binding partners” as defined herein). “Antibody” as used herein is meant to include the entire antibody as well as any antibody fragments capable of binding the antigen or antigenic fragment of interest. Examples of such peptides include complete antibody molecules, antibody fragments, such as Fab, F(ab′)2, CDRS, VL, VH, and any other portion of an antibody which is capable of specifically binding to an antigen. Antibodies for assays of the invention are immunoreactive or immunospecific for, and therefore specifically and selectively bind to, proteins either detected (i.e., biological targets) or used for detection (i.e., probes) in the assays of the invention.
  • The term “binding agent” as used herein refers to any agent that specifically binds to a biological molecule of interest
  • “Complementary” or “substantially complementary” refers to the hybridization or base pairing or the formation of a duplex between nucleotides or nucleic acids, such as, for instance, between the two strands of a double-stranded DNA molecule or between an oligonucleotide primer and a primer binding site on a single-stranded nucleic acid. Complementary nucleotides are, generally, A and T (or A and U), or C and G. Two single-stranded RNA or DNA molecules are said to be substantially complementary when the nucleotides of one strand, optimally aligned and compared and with appropriate nucleotide insertions or deletions, pair with at least about 80% of the other strand, usually at least about 90% to about 95%, and even about 98% to about 100%).
  • “Hybridization” refers to the process in which two single-stranded polynucleotides bind non-covalently to form a stable double-stranded polynucleotide, The resulting (usually) double-stranded polynucleotide is a “hybrid” or “duplex,” “Hybridization conditions” will typically include salt concentrations of approximately less than 1M, often less than about 500 mM and may be less than about 200 mM. A “hybridization buffer” is a buffered salt solution such as 5% SSPE, or other such buffers known in the art. Hybridization temperatures can be as low as 5° C., but are typically greater than 22° C., and more typically greater than about 30° C., and typically in excess of 37° C. Hybridizations are often performed under stringent conditions, i.e., conditions under which a primer will hybridize to its target subsequence but will not hybridize to the other, non-complementary sequences. Stringent conditions are sequence-dependent and are different in different circumstances. For example, longer fragments may require higher hybridization temperatures for specific hybridization than short fragments. As other factors may affect the stringency of hybridization, including base composition and length of the complementary strands, presence of organic solvents, and the extent of base mismatching, the combination of parameters is more important than the absolute measure of any one parameter alone. Generally stringent conditions are selected to be about 5° C. lower than the Tm for the specific sequence at a defined ionic strength and pH. Exemplary stringent conditions include a salt concentration of at least 0.01 M to no more than 1M sodium ion concentration (or other salt) at a pH of about 7.0 to about 8.3 and a temperature of at least 25° C. For example, conditions of 5×SSPE (750 mM NaCl, 50 mM sodium phosphate, 5 mM EDTA at pH 7.4) and a temperature of approximately 30° C. are suitable for allele-specific hybridizations, though a suitable temperature depends on the length and/or GC content of the region hybridized.
  • “Ligation” means to form a covalent bond or linkage between the termini of two or more nucleic acids, e.g., oligonucleotides and/or polynucleotides, in a template-driven reaction. The nature of the bond or linkage may vary widely and the ligation may be carried out enzymatically or chemically. As used herein, ligations are usually carried out enzymatically to form a phosphodiester linkage between a 5′ carbon terminal nucleotide of one oligonucleotide with a 3′ carbon of another nucleotide.
  • “Nucleic acid”, “oligonucleotide”, “oligo” or grammatical equivalents used herein refers generally to at least two nucleotides covalently linked together. A nucleic acid generally will contain phosphodiester bonds, although in some cases nucleic acid analogs may be included that have alternative backbones such as phosphoramidite, phosphorodithioate, or methylphophoroamidite linkages; or peptide nucleic acid backbones and linkages. Other analog nucleic acids include those with bicyclic structures including locked nucleic acids, positive backbones, non-ionic backbones and non-ribose backbones. Modifications of the ribose-phosphate backbone may be done to increase the stability of the molecules; for example, PNA:DNA hybrids can exhibit higher stability in some environments.
  • “Primer” means an oligonucleotide, either natural or synthetic, that is capable, upon forming a duplex with a polynucleotide template, of acting as a point of initiation of nucleic acid synthesis and being extended from its 3′ end along the template so that an extended duplex is formed. The sequence of nucleotides added during the extension process is determined by the sequence of the template polynucleotide. Primers usually are extended by a DNA polymerase.
  • The term “SNP” or “single nucleotide polymorphism” refers to a genetic variation between individuals; e.g., a single nitrogenous base position in the DNA of organisms that is variable. SNPs are found across the genome; much of the genetic variation between individuals is due to variation at SNP loci, and often this genetic variation results in phenotypic variation between individuals. SNPs for use in the present invention and their respective alleles may be derived from any number of sources, such as public databases (U.C. Santa Cruz Human Genome Browser Gateway (http://genome.ucsc.edu/cgi-bin/hgGateway) or the NCBI dbSNP website (http://www.ncbi.nlm.nih.gov/SNP/), or may be experimentally determined as described in U.S. Pat No. 6,969,589; and US Pub. No. 2006/0188875 entitled “Human Genomic Polymorphisms.” Although the use of SNPs is described in some of the embodiments presented herein, it will be understood that other biallelic or multi-allelic genetic markers may also be used. A biallelic genetic marker is one that has two polymorphic forms, or alleles. As mentioned above, for a biallelic genetic marker that is associated with a trait, the allele that is more abundant in the genetic composition of a case group as compared to a control group is termed the “associated allele,” and the other allele may be referred to as the “unassociated allele.” Thus, for each biallelic polymorphism that is associated with a given trait (e.g., a disease or drug response), there is a corresponding associated allele. Other biallelic polymorphisms that may be used with the methods presented herein include, but are not limited to multinucleotide changes, insertions, deletions, and translocations. It will be further appreciated that references to DNA herein may include genomic DNA, mitochondrial DNA, episomal DNA, and/or derivatives of DNA such as amplicons, RNA transcripts, cDNA, DNA analogs, etc. The polymorphic loci that are screened in an association study may be in a diploid or a haploid state and, ideally, would be from sites across the genome.
  • The term “selectively binds”, “selective binding” and the like as used herein, when referring to a binding partner (e.g., protein, nucleic acid, antibody or other affinity capture agent, etc.), refers to a binding reaction of two or more binding partners with high affinity and/or complementarity to ensure selective hybridization under designated assay conditions. Typically, specific binding will be at least three times the standard deviation of the background signal. Thus, under designated conditions the binding partner binds to its particular “target” molecule and does not bind in a significant amount to other molecules present in the sample.
  • “Sequencing”, “sequence determination” and the like means determination of information relating to the nucleotide base sequence of a nucleic acid. Such information may include the identification or determination of partial as well as full sequence information of the nucleic acid. Sequence information may be determined “with varying degrees of statistical reliability or confidence. In one aspect, the term includes the determination of the identity and ordering of a plurality of contiguous nucleotides in a nucleic acid, “High throughput digital sequencing” or “next generation sequencing” means sequence determination using methods that determine many (typically thousands to billions) of nucleic acid sequences in an intrinsically parallel manner, i.e. where DNA templates are prepared for sequencing not one at a time, but in a bulk process, and where many sequences are read out preferably in parallel, or alternatively using an ultra-high throughput serial process that itself may be parallelized. Such methods include but are not limited to pyrosequencing (for example, as commercialized by 454 Life Sciences, Inc., Branford, Conn.); sequencing by ligation (for example, as commercialized in the SOLiD™ technology, Life Technology, Inc., Carlsbad, Calif.); sequencing by synthesis using modified nucleotides (such as commercialized in TruSeq™ and HiSeq™ technology by Illumina, Inc., San Diego, Calif., HeliScope™ by Helicos Biosciences Corporation, Cambridge, Mass., and PacBio RS by Pacific Biosciences of California, Inc., Menlo Park, Calif.), sequencing by ion detection technologies (Ion Torrent, Inc., South San Francisco, Calif.); sequencing of DNA nanoballs (Complete Genomics, Inc., Mountain View, Calif.); nanopore-based sequencing technologies (for example, as developed by Oxford Nanopore Technologies, LTD, Oxford, UK), and like highly parallelized sequencing methods.
  • The term “Tm” is used in reference to the “melting temperature.” The melting temperature is the temperature at which a population of double-stranded nucleic acid molecules becomes half dissociated into single strands. Several equations for calculating the Tm of nucleic acids are well known in the art. As indicated by standard references, a simple estimate of the Tm value may be calculated by the equation, Tm=81.5+0.41 (% G+C), when a nucleic acid is in aqueous solution at 1M NaCl (see e.g., Anderson and Young, Quantitative Filter Hybridization, in Nucleic Acid Hybridization (1985)). Other references (e.g., Allawi and SantaLucia, Jr., Biochemistry, 36:10581-94 (1997)) include alternative methods of computation which take structural and environmental, as well as sequence characteristics into account for the calculation of Tm.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The practice of the techniques described herein may employ, unless otherwise indicated, conventional techniques and descriptions of organic chemistry, polymer technology, molecular biology (including recombinant techniques), cell biology, biochemistry, and sequencing technology, which are within the skill of those who practice in the art. Such conventional techniques include polymer array synthesis, hybridization and ligation of polynucleotides, and detection of hybridization using a label. Specific illustrations of suitable techniques can be had by reference to the examples herein. However, other equivalent conventional procedures can, of course, also be used. Such conventional techniques and descriptions can be found in standard laboratory manuals such as Green, et al., Eds., Genome Analysis: A Laboratory Manual Series (Vols. I-IV) (1999); Weiner, Gabriel, Stephens, Eds., Genetic Variation: A Laboratory Manual (2007); Dieffenbach, Dveksler, Eds., PCR Primer: A Laboratory Manual (2003); Bowtell and Sambrook, DNA Microarrays: A Molecular Cloning Manual (2003); Mount, Bioinformatics: Sequence and Genome Analysis (2004); Sambrook and Russell, Condensed Protocols from Molecular Cloning: A Laboratory Manual (2006); and Sambrook and Russell, Molecular Cloning: A Laboratory Manual (2002) (all from Cold Spring Harbor Laboratory Press); Stryer, Biochemistry (4th Ed.) (1995) W. H. Freeman, New York N.Y.; Gait, “Oligonucleotide Synthesis: A Practical Approach” (2002) IRL Press, London; Nelson and Cox, Lehninger, Principles of Biochemistry (2000) 3rd Ed., W. H. Freeman Pub., New York, N.Y.; and Berg, et al., Biochemistry (2002) 5th Ed., W. H. Freeman Pub., New York, N.Y., all of which are herein incorporated in their entirety by reference for all purposes.
  • Note that as used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “a nucleic acid” refers to one or more nucleic acids, and reference to “the assay” includes reference to equivalent steps and methods known to those skilled in the art, and so forth.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All publications mentioned herein are incorporated by reference for the purpose of describing and disclosing devices, formulations and methodologies that may be used in connection with the presently described invention.
  • Where a range of values is provided, it is understood that each intervening value, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either both of those included limits are also included in the invention.
  • In the following description, numerous specific details are set forth to provide a more thorough understanding of the present invention. However, it will be apparent to one of skill in the art that the present invention may be practiced without one or more of these specific details. In other instances, well-known features and procedures well known to those skilled in the art have not been described in order to avoid obscuring the invention.
  • The Invention in General
  • The assay systems of the invention provide spatially-encoded, multiplexed assays comprising 1) an assay capable of high levels of multiplexing with an efficient spatial encoding scheme; 2) instrumentation capable of delivering reagents according to a spatial pattern; and 3) decoding determined by a readout that is digital in nature. The assay systems of the invention detect the presence or absence and relative amount of a biological target or biological activity indicative of a biological target, as well as the location of the biological target or activity in a biological sample, e.g., a tissue section or other biological structure disposed upon a support such as a microscope slide or culture dish.
  • The assay system further provides instrumentation with an ability to deliver reagents in a spatially-defined pattern. This instrumentation, together “with software, reagents and protocols, provides a key component of the highly innovative assay system of the invention, allowing for measurement of numerous biological targets or activities in a meaningful spatial environment, including gene expression and peptide localization, An encoding scheme used in these assay systems allows one to determine the location of biological targets or activity (or lack thereof) in the biological samples after the products of the multiplexed assay are removed from the biological sample and pooled for analysis. Decoding of the encoding scheme can be performed by, e.g., next-generation sequencing, which easily provides millions to trillions of data points at low cost. The assay results such as the amount or activity of biological targets can then be mapped back to specific location in the biological sample. The assay systems open a new analytical window into the complex spatial patterns of cellular function and regulation in biological samples.
  • A simplified overview of the assay system 100 of the present invention is provided at FIG. 1, At step 110, a biological sample affixed to a support is provided. The biological sample contains biological targets of interest. Biological targets can include any molecule of interest, such as nucleic acids (including, e.g, RNA transcripts, genomic DNA sequences, cDNAs, amplicons, or other nucleic acid sequences) and proteins, enzymes and the like. At step 120, encoded probes are delivered to the biological sample according to a known spatial pattern. Encoded probes comprise probes, which can interact “with biological targets of interest, and coding tags, which identify the positions in the sample of the biological targets being assayed, and thus can be used to link assay results back to locations in the sample. Coding tags in most embodiments are oligonucleotides. However, coding tags may also be mass tags, fluorescent labels, or other moieties.
  • In some embodiments, the probe and coding tag portions of the encoded probe are pre-coupled before being delivered to the biological sample, For example, in the case where the encoded probes are oligonucleotides, both the probe and coding tag sequence can be synthesized as a single oligonucleotide. Alternatively, the probe and coding tag portions of the encoding probes can be synthesized or obtained separately and combined before delivery to the biological sample (e.g., two separate oligonucleotides can be synthesized and coupled by, e.g., ligation; or an antibody and an oligonucleotide can be prepared separately and conjugated before delivery to the biological sample). Also, as is described in FIGS. 2-5, the probes and the coding tags (in encoding oligonucleotides) are synthesized separately, and are delivered to the biological sample at different steps (e.g., probes first and coding tags thereafter, or vice versa) in the assay.
  • At step 130, the encoded probes are allowed to react or interact with the biological targets, i.e., conditions are provided to allow e.g., oligonucleotides to hybridize to nucleic acid targets, enzymes to catalyze reactions with protein targets, antibodies to bind epitopes, etc. In the case where the biological targets are nucleic acids, the encoded probes are typically oligonucleotides and hybridize to the target nucleic acids. In the case that the biological targets are proteins, the encoded probes typically are aptamers, small molecules, or oligonucleotide-conjugated proteins that interact with target proteins by binding to them or by reacting with them (that is, one of the proteins is a substrate for the other). Encoding oligonucleotides may be coupled to the probes (proteins) by conjugation, chemical or photo-crosslinking via suitable groups and the like.
  • Once encoded probes interact with the biological targets, the encoded probes that interacted with the biological targets must be separated from the encoded probes that did not interact with the biological targets at step 140. In the case where the biological targets are nucleic acids and the encoded probes are oligonucleotides, the separation can be accomplished by, e.g., washing the unhybridized encoded probes from the sample. Similarly, for other assays that are based on affinity binding, including those using aptamer, small molecule, and protein probes, washing steps can be used to remove low affinity binders. In the case where the probe is transformed via interaction with the target, e.g., in the case of a peptide, e.g., via cleavage by a protease or phosphorylation by a kinase, it is convenient to collect, all encoded probes—both encoded probes that interacted with the biological targets and were transformed and encoded probes that were not transformed. After collection or pooling, an antibody or other affinity capture agent can be used to capture probes that were transformed by addition of a moiety (e.g., a phosphate group). In cases where probes have been transformed via cleavage, the transformed probes can be separated, e.g., by capturing the non-transformed probes via a tag that is removed from the transformed probes during the transformation (e.g., by cleavage), or by adding a new tag at the site of cleavage.
  • Once the reacted (transformed) or interacted encoded probes are separated from the unreacted or un-interacted encoded probes, the sequence of the reacted and/or interacted encoded probes is determined at step 150 by, preferably, sequencing. The sequence of the encoded probes allows the mapping of the assay results at step 160 back to locations in the biological sample.
  • FIG. 2 provides a simplified overview of an assay system 200 of the present invention embodying an efficient implementation of a combinatorial coding scheme for the encoding of spatial information. For purposes of this overview, the probes are oligonucleotides, but as explained elsewhere, other types of probes can also be used. In step 210, a biological sample affixed to a support, e.g., a tissue sample or other biological structure, is provided. In step 220, one or more oligonucleotide probes are delivered to the biological sample, where the oligonucleotide probes are capable of hybridizing with biological targets in the biological sample. In step 230, the oligonucleotide probes are allowed to interact with (hybridize to) the nucleic acid targets; that is, appropriate conditions are provided where oligonucleotide probes can hybridize to the target nucleic acids.
  • In step 240, the oligonucleotide probes that did not hybridize to target nucleic acids are removed, and thereby separated from oligonucleotide probes that did hybridize to target nucleic acids. In this embodiment, separation can be accomplished by, e.g., washing the sample to remove unhybridized oligonucleotide probes. Next, in step 250, encoding oligonucleotides (the encoding agents) are delivered to the biological sample according to a chosen spatial pattern, where the encoding oligonucleotides comprise coding tags that are used to encode the location of biological targets in the biological sample. Note that in contrast to the assay system of FIG. 1, here the probes and encoding agents (encoding oligonucleotides) are delivered in separate steps. In step 260, the encoding oligonucleotides are coupled to the oligonucleotide probes to create encoded probes. In this case where the probes are oligonucleotides, the encoding oligonucleotides may be coupled to the oligonucleotides probes by, e.g., ligation. Alternatively, the information in the encoding oligonucleotides can be transferred by using a DNA polymerase to extend a probe oligonucleotide that acts as a primer, and thereby copy and incorporate the sequence of the encoding oligonucleotides.
  • In step 270, the sequence of the coding tags in the encoded probes as well as the sequence or a portion of the sequence of the probe itself is determined, and in step 280, the target nucleic acids are mapped back to the biological sample. In some embodiments, the abundance of sequences reveals the relative quantity of biological targets at the location. Although this embodiment shows the individual steps in a particular order, so as to better explain the invention, the precise order of the steps can be varied. For example, steps 220 and 250 can be combined, so that a mixture of the probes and encoding oligonucleotides is delivered according to a chosen spatial pattern. Coupling step 260 can then be carried out immediately after the combined steps 220 and 250, or concomitantly with them. In this case, step 240 would then occur after step 260. It can therefore be appreciated that the two key results of this series of steps, i.e., the location-specific encoding of probe molecules and the separation of probe molecules based on their ability to interact with corresponding target molecules, can be accomplished with some flexibility in the implementation of the particular steps. Similarly, there is considerable flexibility in the design of the coding scheme. As described infra, the assays of the invention are particularly amenable to combinatorial methods.
  • Thus, the present invention provides an ability to look at many different biological targets in many locations, providing the resolution of in situ hybridization with the highly-parallel data analysis of sequencing. In some embodiments, the sum of the multiple biological targets being assayed and the multiple sites in the biological sample is greater than 20, in other embodiments, the sum of the multiple biological targets being assayed and the multiple sites in the biological sample is greater than 50, in other embodiments, the sum of the multiple biological targets being assayed and the multiple sites in the biological sample is greater than 100, greater than 500, 1,000, 10,000, 25,000, 100,000, 500,000, 1,000,000. It will be appreciated that, due to the spatial encoding dimension of the invention, even much larger numbers can be contemplated. For example, assaying 10,000 targets per location×10,000 locations would generate 1O,8 different assays, and even larger numbers than these can easily be contemplated, particularly if spatial locations with resolution on the order of that of single cells are utilized. Further, in embodiments where high-throughput digital sequencing is employed, the sequences of at least 1,000 encoding probes are typically determined in parallel. More typically, using a digital readout, it is desirable to obtain multiple sequence reads for each assay (defined by a probe and a spatial location code). It is desirable to obtain an average of at least 3 copies per assay, and more typically at least 10 or at least 30 copies per assay, depending on the design of the experiment and requirements of the assay. For a quantitative readout with suitable dynamic range, it may be desirable to obtain at least 1,000 reads per assay. Therefore, if 1,000,000 assays are carried out, the number of sequence reads may be 1 billion or more. With high-throughput digital sequencing, and allowing for redundancy, the sequence of at least 10,000 encoding probes are determined in parallel, or the sequence of at least 100,000, 500,000, 1,000,000, 10,000,000, 100,000,000, 1,000,000,000 or more encoding probes are determined in parallel.
  • Assays
  • The assay portion of the assay systems of the present invention comprise the following general steps: delivering probes and encoding agents where the encoding agents (in some embodiments pre-coupled to the probes) are delivered to the sample according to a known spatial pattern, allowing the probes to interact or react with biological targets in the sample, and, if the probes and encoding agents have not been pre- coupled, coupling the encoding agents to probes.
  • The samples of the present invention include virtually any biological sample or samples that can be affixed to a support or provided essentially in a two-dimensional manner, where the ability to tie an assayed biological target or activity back to the location within the biological sample is important. Exemplary biological samples include tissue sections (e.g., including whole animal sectioning and tissue biopsies), cell populations on slides or culture dishes, and the like. The assay systems of the invention are particularly advantageous in that they are compatible with numerous biological sample types, including fresh samples, such as primary tissue sections, and preserved samples including but not limited to frozen samples and paraformalin-fixed, paraffin-embedded (FFPE) samples. An important aspect of the assay systems of the invention is that the biological samples are immobilized on a substrate surface having discrete, independently measurable areas.
  • The biological targets to be detected can be any biological molecules including but not limited to proteins, nucleic acids, lipids, carbohydrates, ions, or multicomponent complexes containing any of the above. Examples of subcellular targets include organelles, e.g., mitochondria, Golgi apparatus, endoplasmic reticulum, chloroplasts, endocytic vesicles, exocytic vesicles, vacuoles, lysosomes, etc.
  • In some particular embodiments, the assay system is used to analyze nucleic acids, e.g., by genotyping, quantitation of DNA copy number or RNA transcripts, localization of particular transcripts within samples, and the like. FIG. 3 illustrates an overall scheme for an exemplary assay for, e.g., detecting single nucleotide polymorphisms (SNPs) that can be used with the assay system of the invention. In FIG. 3, two oligonucleotide probes are provided. Each oligonucleotide probe comprises a target-specific region (located on either side of the SNP to be analyzed) seen at 305 and 307, and ligation regions, seen at 301 and 303, The oligonucleotide probes are allowed to hybridize to a target nucleic acid (not shown) in the biological sample. At step 302, one of the oligonucleotide probes is extended to incorporate the SNP sequence and ligated to the other probe to form an extended probe comprising target nucleic acid region 309 and ligation regions 301 and 303.
  • Two encoding agents, both comprising a coding tag (seen at 315 and 317), a ligation region (seen at 311 and 313), and a primer region (seen at 319 and 321) are combined with and ligated to the extended probe at step 304 to form an encoded target-specific oligonucleotide. Again, in contrast with FIG. 1, the probes and encoding agents are delivered at separate steps. Doing so allows use of the combinatorial embodiments described infra. In preferred embodiments, the encoding oligonucleotides within a pair of encoding oligonucleotides ligate specifically to one side of the target sequence or the other (i.e., 5′ or 3′ of the target sequence) in step 306. Also, typically, the ligation and primer regions of the encoding oligonucleotides and probes are universal; that is, the set of ligation and primer regions used in constructing the probes and encoding oligonucleotides are constant, and only the target-specific regions of the probes and the coding tags of the encoding oligonucleotides differ. However, again in alternative embodiments, the ligation and primer regions are not universal and differ between probes and encoding agents.
  • Following ligation, the encoded probes are eluted, pooled, and, optionally, sequencing adapters are added to the encoded probes via PCR. In alternative embodiments, sequencing primers may be ligated to the encoding oligonucleotides, or sequencing primer sequences can be included as part of the encoding oligonucleotide. As seen in FIG. 3, each sequencing adapter comprises primer region 319 or 321, compatible with the primer regions 319 and 321 on the encoded probes. The final construct comprising first adapter 327, first primer region 319, first coding tag 315, ligation regions 311 and 301, target region 309, ligation regions 313 and 303, second coding tag 317, second primer region 325 and second adapter 329 is now ready for input into a digital high-throughput sequencing process.
  • A combination of extension and ligation reactions are exemplified in FIG. 3, but it should be appreciated that a variety of reactions may be used to couple the encoding oligonucleotides to the target-specific oligonucleotides, including ligation only (e.g., for oligonucleotides that hybridize to contiguous portions of the target nucleic acid sequence). Alternatively, an assay utilizing an additional oligonucleotide, such as in the GOLDENGATE® assay (see Fan, et al., Cold Spring Symp. Quant. Biol., 68:69-78 (2003); (Ilumina, Inc., San Diego, Calif.)), may be employed.
  • In other embodiments, the assay system of the invention also can be used to analyze peptides or proteins, the presence of antibodies, enzymatic and other protein activities, posttranslational modifications, active and non-active forms of peptides, as well as peptide isoforms in a biological sample. Accordingly, the probes may comprise an active region of an enzyme, a binding domain of an immunoglobulin, defined domains of proteins, whole proteins, synthetic peptides, peptides with introduced mutations, aptamers and the like.
  • In certain aspects, the probes are substrates for enzymes or proenzymes, e.g., kinases, phosphatases, zymogens, proteases, or fragments thereof. In certain aspects, the probes are phosphorylation substrates used to detect proteins involved in one or more signal transduction pathways, e.g., a kinase or a phosphatase. In another specific aspect of the invention, the probes are specific protease substrates that associate only with individual proteases or classes of proteases. In other aspects, the probes are different processed forms, isoforms and/or domains of an enzyme. Protein-based probes are typically conjugated or otherwise linked to oligonucleotide encoding agents. The oligonucleotide encoding agents in this case would also include a nucleotide sequence component that allows for identification of the protein probe.
  • In certain aspects, the present invention provides assays for evaluating differences in the amount and/or activity of biological targets between different locations in a sample and/or between samples. The method includes determining a plurality of encoded results from the biological sample and evaluating the differences in quantity of the biological targets at each location in the biological sample.
  • Combinatorial Embodiments
  • To maximize the efficiency of encoding, a combinatorial approach using pairs of coding tags in the encoding oligonucleotides can be used. By de-coupling the target-specific information and the coding tags, the number of oligonucleotides required is dramatically reduced, with a concomitant decrease in cost.
  • FIG. 4 illustrates a general mechanism for one embodiment of a combinatorial encoding scheme of the assay systems of the invention, where nucleic acids in a representative tissue section (shown at 416) are assayed. FIG. 4 at A shows two target-specific/encoding oligonucleotide constructs 420 and 422 (e.g., formed between steps 302 and 304 of FIG. 3) specifically bound to a target nucleic acid 402 of interest, The first encoded probe 420 comprises coding tag 408, associated with, e.g., a universal priming site for amplification of the assay products or an adapter to enable identification of the coding identifiers using sequencing technologies 404. The second encoded probe 422 comprises coding tag 406, associated with, e.g., a universal priming site for amplification of the assay products or an adapter to enable identification of the coding identifiers using sequencing technologies 410.
  • FIG. 4 at B shows the spatial pattern that may be used for twenty different coding tags, a1 through a10 (coding tag 406 on encoded probe 420) and b1 through b10 (coding tag 408 encoded probe 422). Coding tag a1., for example, is deposited on the biological sample in ten discrete areas or spots (shown as the first horizontal line of spots in 412). Coding tag a2 is deposited on the biological sample in ten spots on the second horizontal line in 412. Coding tag a3 is deposited on the biological sample in ten spots on the third horizontal line in 412, and so on. Whereas the “a” tags are deposited in ten horizontal rows, the “b” tags are deposited in ten vertical rows as shown in 414. For example, coding tag b1 is deposited on the biological sample in ten discrete spots in the first vertical row of 414, coding tag b2 is deposited on the biological sample in ten discrete spots in the second vertical row of 414, and so on. Using such a configuration allows for twenty coding tags to uniquely define 100 different locations on the biological sample.
  • FIG. 4 at C shows a representative tissue section 416 coincident with coding tag grid 418. The arrows show how the “a” coding tags and the “b” coding tags are deposited on grid 418 that is coincident with tissue section 416. If, once sequenced, coding tags a1 and b4, e.g., are associated with a target nucleic acid sequence, then that target nucleic acid sequence (i.e., biological target) was present in the tissue section at location a1, b4.
  • FIG. 5 provides a simplified, specific example of the encoding scheme of the assay systems of the invention. FIG. 5 shows encoding oligonucleotides 510, comprising a1, a2, a3, a4 and b1, b3, b3 and b4. Target-specific oligonucleotides (TSOs) (probes) 1 and 2 are shown at 520. A deposit or dispensing scheme is shown at 530. Like the grid exemplified in FIG. 4, encoding oligonucleotides a1 through a4 are deposited in spots in a pattern (here, in a vertical pattern), and encoding oligonucleotides b1 through b4 are deposited in spots in a pattern (here, a horizontal pattern). The grid though shown as a square with spots is actually a deposition pattern on a biological sample (not shown) such as tissue section 416 shown in FIG. 4.
  • The target-specific oligonucleotides are delivered to the biological sample, where the target-specific oligonucleotides hybridize to target nucleic acids in the biological sample if target nucleic acids are present. Unhybridized target-specific oligonucleotides are then removed, e.g., by washing. The encoding oligonucleotides are then delivered to the biological sample according to the spatial pattern shown at 530. The encoding oligonucleotides are ligated (or, e.g., extended and ligated) to any target-specific oligonucleotides that hybridized to the target nucleic acid in the biological sample, the ligated constructs are then eluted from the biological sample, pooled, and sequencing adapters are added through, e.g., PCR or ligation, if the sequences were not previously included in the encoding oligonucleotides. The ligated constructs are sequenced by, e.g., high throughput or “next generation” sequencing.
  • The pool of resulting sequences is shown at 540. A sequence readout was obtained for target-specific oligonucleotide 1 only at a4b1, a4b2, a1b3, a2b3, a3b3, a4b3 and a4b4 (positions shown with horizontal lines). A sequence readout was obtained for target-specific oligonucleotide 2 only at a1b1 (position shown with vertical lines). A sequence readout was obtained for both target- specific oligonucleotides 1 and 2 at positions a2b1, a3b1, a1b2, a2b2, and a3b2 (positions shown with cross-hatching). No sequence readout was obtained for either target-specific oligonucleotides at a1b4, a2b4 or a3b4 (positions shown without shading). Thus, in the biological sample on which the assay took place the first target nucleic acid was detected in a large portion of the left side and at the bottom of the biological sample, the second target nucleic acid was detected only in the upper left portion of the biological sample, and neither target nucleic acid was detected in the upper right portion of the biological sample. The differential expression of the two target nucleic acids now can be mapped back to the biological sample and to the biological structures or cell types in these locations in the biological sample, as shown in 550.
  • In addition to location information, information relating to relative abundance of the encoded tags can be obtained. For example, if it is found that there are ten times as many a4T1b1 sequences occurring in the data set as compared to a4T1b2 sequences, this would indicate that target nucleic acid sequence 1 is ten times more abundant at the a4T1b1 location than at the a4T1b2 location.
  • In the case of nucleotide analysis as shown in FIG. 3, by ligating the coding tags directly to target-specific oligonucleotides, only 2n target-specific oligonucleotides are needed for n targets. For example, using the combinatorial approach outlined in FIG. 2, assaying 100 different targets at 10,000 spatial locations would require 2×100 target-specific oligonucleotides and 2×100 encoding oligonucleotides. The total count of assay oligonucleotides would be only 400 (200 target-specific and 200 encoding), not counting universal primers. In contrast, if the coding oligonucleotides were not decoupled from the target-specific oligonucleotides, (n×X positional codes)+(n×Y positional codes) would be needed, or in the above example, 20,000 oligonucleotides, not counting universal primer sequences. Moreover, though the embodiments shown in FIGS. 2-5 depict a combinatorial scheme using two encoding agents (coding tags), three, four or more encoding agents and coding tags may be used, and attached to the probe or one another by varying means and in varying combinations of steps.
  • Due to the spatial encoding aspect of the assay system of the invention, a large amount of information can be generated with even a modest number of assays. For example, five or more biological targets assayed at five or more positions in the sample generates 25 or more combinations. Using digital sequencing as a readout, the optimum number of sequence reads per combination depends on the sensitivity and dynamic range required, and can be adjusted, For example, if for each combination on average 100 reads are sampled, the total for 25 combination is 25,000 reads. If 1,000 targets are assayed at 1,000 locations with an average sampling depth of 1,000, then 109 reads are required. These numbers, although large, are within the capacity of intrinsically parallel digital sequencing methods, which can generate datasets of billions or even trillions of reads in a reasonable timeframe and at a very low cost per read. Therefore, by varying the numbers of positions interrogated or biological targets assayed, or both, and using digital sequencing, large amounts of information can be obtained. In specific aspects, multiple locations are interrogated for two or more biological molecules.
  • Reagent Delivery Systems
  • The reagent delivery system of the invention includes instrumentation that allows the delivery of reagents to discrete portions of the biological sample, maintaining the integrity of the spatial patterns of the encoding scheme. Reagent delivery systems of the assay systems of the invention comprise optional imaging means, reagent delivery hardware and control software. Reagent delivery can be achieved in a number of different ways. It should be noted that reagent delivery may be to many different biological samples at one time. A single tissue section has been exemplified herein; however, multiple biological samples may be affixed and analyzed simultaneously. For example, pions of a tissue sample can be analyzed in parallel and the data combined to build a 3D map.
  • Integral to the assay system of the invention is instrumentation that allows for spatial patterning of reagents onto the biological sample. Technologies for formulating and delivering both biological molecules (e.g. oligonucleotides or antibodies) and chemical reagents (e.g., small molecules or dNTPs) are known in the art, and uses of these instrument systems are known to one skilled in the art and easily adaptable to the assay systems of the invention. One example of a suitable reagent delivery system is the LabcyteTM Echo acoustic liquid handier, which can he used to deliver nanoliter scale droplets containing biological molecules with high precision and reproducibility. One skilled in the art could incorporate this reagent delivery device into the overall system, using software to specify the locations to which reagents should be delivered.
  • Other instruments that can be used for the deposition of agents and/or coding identifiers onto biological samples include, but are not limited to, ink jet spotting; mechanical spotting by means of pin, pen or capillary; micro contact printing; photochemical or photolithographic methods; and the like. For several applications, it may be preferred to segment or sequester certain areas of the biological samples into one or more assay areas for different reagent distributions and/or biological target determination.
  • The assay areas may be physically separated using barriers or channels.
  • In one exemplary aspect, the reagent delivery system may be a flow-based system. The flow-based systems for reagent delivery in the present invention can include instrumentation such as one or more pumps, valves, fluid reservoirs, channels, and/or reagent storage cells. Reagent delivery systems are configured to move fluid to contact a discrete section of the biological sample. Movement of the reagents can be driven by a pump disposed, for example, downstream of the fluid reagents. The pump can drive each fluid reagent to (and past) the reaction compartment. Alternatively, reagents may be driven through the fluid by gravity. US Pub. Nos. 20070166725 and 20050239192 disclose certain general-purpose fluidics tools that can be used with the assay systems of the invention, allowing for the precise manipulation of gases, liquids and solids to accomplish very complex analytical manipulations with relatively simple hardware.
  • In a more specific example, one or more flow-cells can be attached to the substrate-affixed biological sample from above. The flow-cell can include inlet and outlet tubes connected thereto and optionally an external pump is used to deliver reagents to the flow-cell and across the biological sample. The flow cells are configured to deliver reagents only to certain portions of the biological sample, restricting the amount and type of reagent delivered to any specific section of the biological sample.
  • In another aspect, a microfluidic system can be integrated into the substrate upon which the biological sample is disposed or externally attached on top of the substrate. Microfluidic passages for holding and carrying fluid may be formed on and/or above the planar substrate by a fluidics layer abutted to the substrate. Fluid reagents can be selected and delivered according to selective opening and closing of valves disposed between reagent reservoirs.
  • Pumps generally include any mechanism for moving fluid and/or reagents disposed in fluid. In some examples, the pump can be configured to move fluid and/or reagents through passages with small volumes (i.e., microfluidic structures). The pump can operate mechanically by exerting a positive or negative pressure on fluid and/or on a structure carrying fluid, electrically by appropriate application of an electric field(s), or both, among other means. Exemplary mechanical pumps may include syringe pumps, peristaltic pumps, rotary pumps, pressurized gas, pipettors, etc. Mechanical pumps may be micromachined, molded, etc. Exemplary electrical pumps may include electrodes and may operate by electrophoresis, electroendoosmosis, electrocapillarity, dielectrophoresis (including traveling wave forms thereof), and/or the like.
  • Valves generally include any mechanism for regulating the passage of fluid through a channel. Valves can include, for example, deformable members that can be selectively deformed to partially or completely close a channel, a movable projection that can be selectively extended into a channel to partially or completely block a channel, an electrocapillary structure, and/or the like.
  • An open gasket can be attached to the top of the biological sample and the sample and reagents can be injected into the gasket. Suitable gasket materials include, but are not limited to, neoprene, nitrile, and silicone rubber. Alternatively, a watertight reaction chamber may be formed by a gasket sandwiched between the biological sample on the substrate and a chemically inert, water resistant material such as, but not limited to, black-anodized aluminum, thermoplastics (e.g., polystyrene, polycarbonate, etc), glass, etc.
  • In an optional embodiment, the assay system comprises imaging means to determine features and organization of the biological sample of interest. The images obtained, e.g., may be used to design the deposition pattern of the reagents, Imaging means are optional, as an individual can instead view the biological sample using, e.g., a microscope, analyze the organization of the biological sample, and specify a spatial pattern for delivery assay reagents. If included, the delivery system can comprise a microcircuit arrangement including an imager, such as a CCD or IGFET-based (e.g., CMOS-based) imager and an ultrasonic sprayer for reagent delivery such as described in US Pub. No. 20090197326, which is incorporated herein by reference. Also, it should be noted that although FIGS. 4 and 5 illustrate using a x,y grid configuration, other configurations can be used, such as, e.g., following the topology of a tissue sample; targeting certain groups of cells, cell layers and/or cell types in a tissue, and the like.
  • In yet another alternative, the reagent delivery system controls the delivery of reagents to specific patterns on a biological sample surface using semiconductor techniques such as masking and spraying. Specific areas of a biological sample can be protected from exposure to reagents through use of a mask to protect specific areas from exposure. The reagents may be introduced to the biological sample using conventional techniques such as spraying or fluid flow. The use of masked delivery results in a patterned delivery scheme on the substrate surface.
  • In a preferred aspect of the invention, the reagent delivery instrumentation is based on inkjet printing technology. There are a variety of different ink-jetting mechanisms (e.g., thermal, piezoelectric) and compatibility has been shown with aqueous and organic ink formulations. Sets of independently actuated nozzles can be used to deliver multiple reagents at the same time, and very high resolutions are be achieved.
  • In order to target specific sites of interest, an informative image of the biological sample to be assayed may be used to assist in the reagent delivery methods and associated encoding scheme. Sample regions of the biological sample can be identified using image processing (e.g., images of cell types differentiated by immunohistochemistry or other staining chemistries) integrated with other features of the assay system. In some aspects, software is used to automatically translate image information into a reagent delivery pattern. A mechanism to register and align very precisely the biological sample for reagent delivery is thus an important component of the assay systems of the invention. Mechanisms such as the use of fiducial markers on slides and/or other very accurate physical positioning systems can be adapted to this purpose.
  • The invention preferably comprises a complete suite of software tailored to the assay system. Optionally, oligonucleotide design software is used to design the encoding nucleotides (and in embodiments where nucleic acids are assayed, the target-specific oligonucleotides) for the specific assay to be run, and may be integrated as a part of the system. Also optionally, algorithms and software for reagent delivery and data analysis (i.e., sequence analysis) may be integrated to determine assay results. Integrated data analysis is particularly useful, as the type of dataset that is generated may be massive as a consequence of scale. Algorithms and software tools that are specifically designed for analysis of the spatially-associated data generated by the assay systems, including pattern-analysis software and visualization tools, enhance the value of the data generated by the assay systems.
  • In certain aspects, the assay system comprises processes for making and carrying out the quality control of reagents, e.g., the integrity and sequence fidelity of oligonucleotide pools. In particular, reagents are formulated according to factors such as volatility, stability at key temperatures, and chemical compatibility for compatibility with the reagent delivery instrumentation and may be analyzed by instrumentation integrated within the assay system.
  • Sequencing
  • Numerous methods can be used to identify the coding tags and probe sequences in the encoded probes of the assay systems of the invention. The coding tags can be detected using techniques such as mass spectroscopy (e.g., Maldi-T of, LC-MS/MS), nuclear magnetic resonance imaging, or, preferably, nucleic acid sequencing. Examples of techniques for decoding the coding tags of the present invention can be found, for example, in US Pub. No. 20080220434, which is incorporated herein by reference. For example, the coding tags may be oligonucleotide mass tags (OMTs or massTags). Such tags are described, e.g., in US Pub. No. 20090305237, which is incorporated by reference in its entirety. In yet another alternative, the encoded probes can be amplified and hybridized to a microarray. This would require separate amplification reactions to be carried out, in which each amplification is specific to a particular spatial code or subset of codes, accomplished by using code-specific primers. Each amplification would also incorporate a different resolvable label (e.g. fluorophor). Following hybridization, the relative amounts of a particular target mapping to different spatial locations in the sample can he determined by the relative abundances of the resolvable labels.
  • In one particularly preferred aspect, the resulting coding tags according to the assay system are substrates for high-throughput, next-generation sequencing, and highly parallel next-generation sequencing methods are used to confirm the sequence of the coding tags, for example, with SOLiD™ technology (Life Technologies, Inc.) or Genome Ananlyzer (Illumina, Inc.). Such next-generation sequencing methods can be carried out, for example, using a one pass sequencing method or using paired-end sequencing. Next generation sequencing methods include, but are not limited to, hybridization-based methods, such as disclosed in e.g., Drmanac, U.S. Pat. Nos. 6,864,052; 6,309,824; and 6,401,267; and Drmanac et al, U.S. patent publication 2005/0191656; sequencing-by-synthesis methods, e.g., U.S. Pat Nos. 6,210,891; 6,828,100; 6,969,488; 6,897,023; 6,833,246; 6,911,345; 6,787,308; 7,297,518; 7,462,449 and 7,501,245; US Publication Application Nos. 20110059436; 20040106110; 20030064398; and 20030022207; Ronaghi, et al, Science, 281: 363-365 (1998); and Li, et al, Proc. Natl. Acad. Sci., 100: 414-419 (2003); ligation-based methods, e.g., U.S. Pat. Nos. 5,912,148 and 6,130,073; and U.S. Pat. Appln Nos. 20100105052, 20070207482 and 20090018024; nanopore sequencing e.g., U.S. Pat. Appln Nos. 20070036511; 20080032301; 20080128627; 20090082212; and Soni and Meller, Clin Chem 53:1996-2001 (2007)), as well as other methods, e.g., U.S. Pat. Appln Nos. 20110033854; 20090264299; 20090155781; and 20090005252; also, see, McKernan, et al., Genome Res., 19:1527-41 (2009) and Bentley, et al., Nature 456:53-59 (2008), all of which are incorporated herein in their entirety for all purposes.
  • Applications of Assay System
  • It will be apparent to one skilled in the art upon reading the present disclosure that there are numerous important areas of biological research, diagnostics, and drug development that will benefit from a high throughput multiplexed assay system that can measure simultaneously the amount and spatial location of a biological target in a biological sample. For example, combining the ability to estimate the relative abundance of different RNA transcripts with the ability to reconstruct an image of spatial patterns of abundance across many locations, which may be as small as or even smaller than individual cells, in a tissue enables many different areas of basic research. The following are exemplary uses and are by no means meant to be limiting in scope.
  • In one example, 3-dimensional patterns of gene expression are determined by analyzing a series of tissue sections, in a manner analogous to image reconstruction in CT scanning, Such a method can be used to measure changes in gene expression in disease pathology, e.g., in cancerous tissue and/or a tissue upon injury, inflammation or infection. With the assay systems of the invention, more detailed information on gene expression and protein localization in complex tissues is obtained, leading to new insights into the function and regulation both in normal and diseased states, and provides new hypotheses that can be tested. For example, an assay system of the invention may enable some of the insights gained from many individual studies and larger programs like ENCODE (Birney, et al., Nature, 447:799-816 (2007)) and modENCODE to be integrated at the tissue level. The assay systems also aid computational efforts to model interacting networks of gene expression in the field of systems biology.
  • The assay systems also provide a novel approach to analysis of somatic variation, e.g., somatic mutations in cancer or variability in response to infectious organisms. For example, tumors are typically highly heterogeneous, containing cancer cells as well as genetically normal cells in an abnormal local environment. Cancer cells undergo mutation and selection, and in this process it is not unusual for local clones to develop. Identifying relatively rare somatic mutations in the context of tumors may enable the study of the role of key mutations in the selection of clonal variants. Transcriptional patterns associated with angiogenesis, inflammation, or other cancer-related processes in both cancer and genetically normal cells can be analyzed for insights into cancer biology and assist in the development of new therapeutic agents for the treatment of cancers. In another example, individuals have varying susceptibility to infectious organisms, and the assay systems of the invention can be used to study the interaction between microbes and tissues or the various cell types within the tissue.
  • Importantly, in addition to providing spatially-associated information, the invention allows a great increase in the sensitivity of detecting rare mutations, as signal to noise can he dramatically increased since only a small location is assayed in any given reaction. In a typical assay for rare mutations in a mixed sample, the sample is treated in bulk, i.e., nucleic acids are extracted from many cells into a single pool. Thus, if a mutation is present in one cell in 10,000, it must be detected against a background of normal DNA from 10,000 cells. In contrast, with the assay systems of the invention many cells can be analyzed, but individual cells or small groups of cells would be identified by the spatial coding system. Therefore, in the assay systems of the present invention, background is reduced by orders of magnitude, greatly increasing sensitivity. Furthermore, the spatial organization of mutant cells can be observed, which may be particularly important in detecting key mutations in tissue sections in cancer. Already molecular histological analyses are yielding insights into cancer biology and may have potential for use in diagnostics. The technology of the invention promises to greatly increase the power of such approaches.
  • The present invention provides assays, assay systems, and methods of using such assays in spatially encoded biological assays. The invention provides an assay system comprising one or more agents provided in defined spatial patterns on a substrate surface, and a detection system for identifying the presence or absence, relative amount, and location of a biological molecule. Such biological molecules include, but are not limited to, nucleic acids, peptides, carbohydrates, cellular components, and the like. The assay system is a novel multiplexing approach, as it allows multiple molecules and their respective multiple locations to be identified in a single system using a unique encoding scheme. This encoding scheme uses both molecule-specific binding agents and coding identifiers to provide a practical and cost-effective determination of information on multiple biological molecules, including specific positional information of such molecules in a biological sample, e.g., a tissue section. The single molecule detection analysis using the encoding system also allows relative amounts of biological molecules to be detected, thus providing information on expression levels, sequestering in specific locales, and the like.
  • The assay systems detect the presence or absence, and relative amount, of a biological molecule at more than one spatial location in a sample. In addition, the assays provide methods for doing this for multiple biological molecules simultaneously. The assay systems utilize one or more binding agents that specifically bind to the biological molecule of interest and unique coding identifiers associated with specific binding agents. The detection system utilizes a method for identifying the presence and spatial address of the agent binding based on the positive and/or negative results that are obtained using detection of the agent and identifier and the encoding scheme of the spatial patterns on the substrate surface. In a specific aspect, the encoding scheme employs limited reagent delivery to the spatial patterns on the substrate surface, and access of the coding identifiers and/or binding agents to portions of the sample is controlled through such limited delivery.
  • In one aspect, the assay system detects the presence or absence and spatial location of a biological molecule based on the positive and/or negative results that are obtained using limited reagent delivery and the encoding scheme of the spatial patterns on the substrate surface.
  • The assay system and methods of the invention are based on relational, solid-state substrates with positions that represent specific spatial locations within a biological sample, e.g., a cell, organelle or tissue. The ability to use encoding features to represent locations allows high-throughput analysis of the presence or absence, and relative amount, of a biological molecule at more than one spatial location in a sample. The encoding features also allow provide assaying of multiple biological molecules at these multiple locations simultaneously.
  • A primary feature of the invention is the preservation of the spatial organization of elements in a sample of interest through the use of an encoding scheme. For example, the assay may be designed to preserve the relative position of cells in a tissue, and the assay may interrogate the individual cells for genomic DNA variation (including epigenetic modifications), and RNA and protein expression.
  • In one specific aspect, the encoding scheme of the assay system comprises the use of two or more coding patterns, each comprising regions defined by spatial patterns on the substrate surface. For example, the assay system can utilize an encoding scheme that comprises a 2-dimensional grid format based on the discrete positioning of the binding agents in the substrate surfaces. In another example, the spatial patterns may be based on more randomized cell locations, e.g., the patterns on the substrate surface follow an underlying biological structure rather than a strict, x,y grid pattern. This aspect includes systems with two or more substantially identical spatial patterns using different binding agents and/or coding identifiers, as well as systems having different patterns for different agents and/or coding identifiers. The encoding scheme of the systems can be controlled by delivery of different reagents to discrete regions on the substrate surfaces, which allows different reactions to take place on substantially similar agents of known location on the substrate surfaces.
  • In one specific aspect, the invention provides high resolution, high- throughput analysis of nucleic acids and/or expression levels that provides both detection and spatial identification of large numbers of nucleic acids, e.g., DNA or RNA.
  • In another specific aspect, the invention provides high resolution, high- throughput analysis of proteins that provides both detection and spatial identification of large numbers of such proteins, e.g., kinases or proteases.
  • Numerous reagent delivery systems can be used with the assay system of the invention. The primary criteria of such reagent delivery systems is the ability to direct delivery of specific agents based on spatial patterns on the substrate surface.
  • In one preferred aspect, the encoding scheme utilizes a reagent delivery system based on printing and informatics technologies to implement the spatial patterns used for identification and localization of the biological materials. For example, the patterns found in the encoding scheme may be created using ink jet printing technology to provide reagents at specific locations on one or more substrate surfaces. The desired patterns are set out in specific coding patterns on the substrate surface.
  • In certain aspects of the invention, the binding agents are immobilized directly to the substrate surface, and the location of the binding agents is known or determined prior to use of the substrate surface in the assay system. In another aspect, the binding agents are immobilized onto beads or other separate structural elements that are then provided in known locations on the substrate surface. In yet another aspect, the binding agents may be provided in or on features of the substrate surface, e.g., provided in wells or channels.
  • In specific aspects of the invention, the binding agents are nucleic acids immobilized directly or indirectly to the substrate surface, e.g., directly through the use of amino groups on the substrate surface or indirectly through the use of a linker. The location of the nucleic acid sequences is known or determined prior to use of the substrate surface in the assay system. In another specific aspect, the nucleic acids may be immobilized directly or indirectly onto beads that are then provided in known locations on the substrate surface. In yet another aspect, the nucleic acids may be provided in or on features of the substrate surface, e.g., provided in wells.
  • In these aspects involving nucleic acid agents, any methods of sequence determination can be used, e.g., sequencing, hybridization and the like. In a preferred aspect, nucleic acid sequencing, and preferably next-generation sequencing, is used to decode the spatial encoding scheme in the assay system of the invention. This provides a very wide dynamic range for very large numbers of assays, allowing for efficient multiplexing.
  • In some aspects, the assay utilizes two or more oligonucleotides, the oligonucleotides comprising a universal primer region and a region that correlates specifically to a single spatial pattern within the spatial encoding scheme. In a specific aspect, the assay comprises two allele specific oligonucleotides and one locus specific oligonucleotides. These oligonucleotides allow the identification of specific SNPs, indels or mutations within an allele. This is useful in the identification of genetic changes in somatic cells, genotyping of tissues, and the like.
  • In other specific aspects of the invention, the binding agents are peptides. In one aspect, these peptides are associated directly or indirectly to known locations on a substrate surface, e.g., using binding protein pairs or through oligonucleotide linkers complementary to oligonucleotides on the substrate surface. In another aspect, the binding agents are peptides are immobilized directly or indirectly onto beads or other separate structural elements that are then provided in known locations on the substrate surface. In yet another aspect, the peptides may be provided in or on features of the substrate surface, e.g., provided in wells.
  • In yet other specific aspects of the invention, the binding agents are chemical entities (e.g., small molecules) that are coded, e.g. using sequence tags or mass spectroscopy tags as coding identifiers. In one aspect, these chemical entities can be are immobilized directly to the substrate surface. In another aspect, the binding agents are immobilized onto beads or other separate structural elements that are then provided in known locations on the substrate surface. In yet another aspect, the binding agents may be provided in or on features of the substrate surface, e.g., provided in wells.
  • The assay system of the invention can utilize various detection mechanisms, based on the molecules to be detected and the reagents needed for such detection system. Exemplary methods that can be used with the assay systems of the invention are described in more detail below.
  • The Invention in General
  • The assay system and methods of the invention are based on relational methods that allow extraction of data to detect the presence or absence and relative amount of a biological molecule, and the location of this molecule in a sample having a distinct structure, e.g., a tissue section or other biological structure with distinct locations of specific biological molecules. The encoding scheme used in these systems corresponds to the structural elements of the sample, and the information obtained using a two-dimensional coding system is indicative of the spatial addresses of these molecules in a sample of interest.
  • Integral to the assay system of the invention is a method for spatial patterning of reagents. Technologies for formulating and delivering both biological molecules (e.g. DNA or antibodies) and chemical reagents (e.g., small molecules or dNTPs) have already been demonstrated, and use of these systems will be available to one skilled in the art and easily adaptable upon reading this specification.
  • The assay design of the invention provides an accurate and easily scalable spatial encoding system. The ability to deliver reagents in a spatially defined pattern together with software, reagents and protocols comprises a novel and highly innovative assay system for spatial analysis of various biological molecules and activities. This allows the assays to measure numerous biological functions in a meaningful spatial environment, including functions such as gene expression and peptide localization. The systems provide the potential to open a new analytical window into the complex spatial patterns of cellular function and regulation in biological systems.
  • The biological molecules to be detected can be any biological molecules such as proteins, nucleic acids, lipids, carbohydrates, ions, or multicomponent complexes containing any of the above. Further examples of subcellular objects include organelles, e.g., mitochondria, Golgi apparatus, endoplasmic reticulum, chloroplast, endocytic vesicle, exocytic vesicles, vacuole, lysosome, etc.
  • FIG. 1 illustrates such a target-specific assay system for identification of nucleic acid sequences in a sample. In this system 101, two reagents 120, 122 that specifically bind to a biological molecule of interest are associated with coding identifiers 106, 108 that encode for a spatial location in the sample. These coding identifiers 106, 108 are optionally associated with sites that assist in their identification in the assay format, e.g., universal priming sites 104, 110 for amplification of the assay products or adapters to enable identification of the coding identifiers and the binding agents using sequencing technologies. The sample that is tested, here shown as a tissue section 116 is encoded using the combination of the patterns 112, 114 created using the separate coding identifiers 106, 108 which provide a two dimensional code 118 that shows the location of any positive detection of the biological molecule 102 as well as quantifying the biological molecule 102 at each location assayed in the tissue.
  • The assay systems of the invention are particularly advantageous in that they are compatible with numerous samples types, such as fresh samples, such as primary tissue sections, and preserved samples including but not limited to frozen samples and paraformalin-fixed, paraffin-embedded (FFPE) samples. An important aspect of the assay systems of the invention is that the binding agents are immobilized on a substrate surface in discrete, independently measureable areas. These discrete areas can be formed by spatially selective deposition of the binding agents on the substrate surface. Numerous methods can be used for the deposition of the agent and the coding identifiers associates with the agent. For example, the coding identifiers can be delivered together or separately from the agent. If delivered together they can be attached (e.g., synthesized as a single molecule or attached through ligation or a chemical coupling mechanism) or simply mixed together to be attached after delivery to the substrate. In a preferred aspect, the agent and the coding identifier are made separately, mixed together for attachment, and delivered either attached or as a mixture to be attached on the surface. In a specific aspect the binding agents are delivered generally over the substrate surface and the coding identifiers are delivered in a pattern-specific manner.
  • Examples of methods that can be used for deposition of agents and/or coding identifiers onto the substrate surface include, but are not limited to, ink jet spotting, mechanical spotting by means of pin, pen or capillary, micro contact printing, fluidically contacting the measurement areas with the biological or biochemical or synthetic recognition elements upon their supply in parallel or crossed micro channels, upon exposure to pressure differences or to electric or electromagnetic potentials, and photochemical or photolithographic immobilization methods.
  • For several applications, it may be preferred to arrange the substrates into segments of one or more measurement areas for reagent distribution and agent determination. These regions may be physically separated using barriers or channels. They may still comprise several additional discrete measurement areas with agents that are different or in different combination from each other.
  • In certain aspects, the present invention provides a method, e.g., a machine-based method, for evaluating changes in the presence and/or location of a biological molecule over time. The method includes providing a plurality of encoded array results representative of the biological molecule over time and evaluating the differences in detection and/or localization of the biological molecules.
  • Nucleic Acid Detection and Localization
  • In a particular aspect, the assay system is used to analyze nucleic acids, e.g genotyping, gene expression analysis, localization of particular transcripts within samples, and the like.
  • Genotyping may be performed using any technique known to those of skill in the art. Preferred techniques permit rapid, accurate determination of multiple variations with a minimum of sample handling. Some examples of suitable techniques involve but are not limited to direct DNA sequencing, capillary electrophoresis, hybridization, allele-specific probes or primers, single-strand conformation polymorphism analysis, nucleic acid arrays, bead arrays, restriction fragment length polymorphism analysis, cleavage fragment length polymorphism analysis, random amplified polymorphic DNA, ligase detection reaction, heteroduplex or fragment analysis, differential sequencing with mass spectrometry, atomic force microscopy, pyrosequencing, FRET (e.g., TaqMan (Applied Biosystems, Inc., Foster City, Calif.) and Molecular Beacon (Stratagene, La Jolla, Calif.) assays), and other related techniques. Several methods for DNA sequencing are well known and generally available in the art. See, for example, Sambrook, et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory, New York) (2001); Ausubel, et al., Current Protocols in Molecular Biology (John Wiley and Sons, New York) (1997), Twyman, et al. (2003) “Techniques Patents for SNP Genotyping”, Pharmacogenomics 4(1):67-79; and Kristensen, et al. (2001) “High-Throughput Methods for Detection of Genetic Variation”, BioTechniques 30(2):318-332. For details on the use of nucleic acid arrays (DNA chips) for the detection of, for example, SNPs, see U.S. Pat. No. 6,300,063 issued to Lipshultz, et al., and U.S. Pat. No. 5,837,832 to Chee, et al., HuSNP Mapping Assay, reagent kit and user manual, Affymetrix Part No. 90094 (Affymetrix, Santa Clara, Calif). The molecular inversion probe (MIP) assay format (Hardenbol et al., 2003) is another example of a highly multiplexable assay that may be used with the assay systems of the invention.
  • In one exemplary and preferred method for analyzing nucleic acids using the assay system of the invention, the detection of nucleic acids uses two allele-specific oligonucleotides and a locus specific oligonucleotide. The assay methods are carried out according to the strategy outlined in FIG. 2 using next-generation sequencing or another highly parallel nucleic acid assay technology. In this assay, a set of two oligonucleotides is designed to hybridize to each target sequence, with a common oligonucleotide and two unique coding identifiers. The allele can be determined, e,g, by primer extension of the locus specific oligonucleotide. Following primer extension and ligation, an amplifiable template is formed with universal primer sequences at either end. Assay oligonucleotides are annealed to a template and enzymatic reactions are used to join the two oligonucleotides only when both are correctly annealed. The detection techniques and read out parameters used in this system of the invention include a much shorter tag than the oligonucleotides used in the assays that are based on capture by hybridization. These shorter tags are designed to be read out by sequencing or, preferably, used to ligate codes onto both ends of the fragment as illustrated in FIG. 2.
  • In FIG. 2, two target-specific assay oligonucleotides are ligated together 202 following in situ hybridization to target sequences. At the same time, encoding oligonucleotides containing tag sequence sets X and Y are ligated 204 to the target specific oligonucleotides. Oligonucleotides containing X ligate specifically to one side of the targeting construct and oligonucleotides containing Y ligate to the other. The oligonucleotides contain universal primer sites P1 and P2. Following ligation, the constructs are eluted and, optionally, sequencing adapters can be attached 206, e.g., via PCR.
  • In one preferred aspect, the final construct created from the assay method is a substrate for next-generation sequencing, and highly parallel next-generation sequencing methods are used to confirm the sequence of constructs. Such sequencing methods can be carried out, for example, using a one pass sequencing method or using paired-end sequencing. Next generation sequencing methods include, but are not limited to, hybridization-based methods, such as disclosed in Drmanac, U.S. Pat. Nos. 6,864,052; 6,309,824; and 6,401,267; and Drmanac et al, U.S. patent publication 2005/0191656, and sequencing by synthesis methods, e.g., Nyren et al, U.S. Pat. No. 6,210,891; Ronaghi, U.S. Pat. No. 6,828,100; Ronaghi et al (1998), Science, 281: 363-365; Balasubramanian, U.S. Pat. No. 6,833,246; Quake, U.S. Pat. No. 6,911,345; Li et al, Proc. Natl. Acad. Sci., 100: 414-419 (2003); Smith et al, PCT publication WO 2006/074351; use of reversible extension terminators, e.g., Turner, U.S. Pat. No. 6,833,246 and Turner, U.S. Pat. No. 6,833,246 and ligation-based methods, e.g., Shendure et al (2005), Science, 309: 1728-1739, Macevicz, U.S. Pat. No. 6,306,597; which references are incorporated by reference. Soddart et al., PNAS USA. 2009 Apr. 20; Xiao et al., Nat Methods. 2009 March;6(3):199-201. Epub 2009 Feb. 8.
  • To maximize the efficiency of encoding, a combinatorial approach using pairs of oligonucleotides can be used. For example, with only two sets of 100 codes, a substrate can theoretically encode up to 10,000 locations. The number of assay oligonucleotides required is dramatically reduced, the cost decreased, and the robustness of the approach increased by decoupling the coding sequences from the genome-specific sequences. Alternative assay formats can also be used (e.g. ligation or primer extension followed by ligation).
  • By ligating the codes on separately, only 2n target-specific assay oligonucleotides are needed for n targets. For example, assaying 100 different targets at 10,000 spatial locations would require 2×100 targeting oligonucleotides and 2×100 encoding oligonucleotides, using a combinatorial approach outlined in FIG. 2. The total count of assay oligonucleotides would be only 400 (200 target-specific and 200 encoding), not counting universal primers. In contrast, if the coding oligonucleotides were not decoupled, (n×X positional codes)+(n×Y positional codes) would be needed, or in the above example, 20,000 oligonucleotides, not counting universal primer sequences.
  • Due to the matrix system of the invention, a large amount of information can be obtained even using five or more positions interrogated for five or more biological molecules. By varying one or the other of these, large amounts of information can be obtained, both in terms of locations and/or specific biological In specific aspects, multiple locations are interrogated for two or more biological molecules. As an example, for each datapoint ˜1,000 reads may be sampled, for a total of −10E9 reads for 10E6 datapoints.
  • Peptide Detection Systems
  • The assay system of the invention can be used to analyze biological molecules using peptide agents that are associated with the substrate surface in a spatial pattern. Such peptides may comprise an active region of an enzyme, a binding domain of an immunoglobulin, defined domains of proteins, whole proteins, synthetic peptides, peptides with introduced mutations, etc.
  • The assay system of the invention allows the identification and spatial location of various forms of peptides, including isoforms and peptides that have undergone posttranslational modification. Importantly, certain aspects of the invention allow the identification of active versus non-active forms of such peptides in a sample. This allows the identification of the presence or absence of specific peptide isoforms, and also acts as a proxy for identification of peptide activity in a sample.
  • In certain aspects of the invention, the binding agents associated with the substrate surfaces of the assay system include substrates for enzymes or proenzymes, e.g., a kinase, a phosphatase, a zymogen, a protease, or a fragment thereof. In certain aspects, the binding agents associated with the substrate surfaces are phosphorylation substrates used to detect proteins involved with one or more signal transduction pathways, e.g., a kinase or a phosphatase. In another specific aspect of the invention, the binding agents are specific protease substrates that associate only with individual or classes of proteases. In other aspects, the binding agents on the substrate surface are different processed forms, isoforms and/or domains of an enzyme.
  • Reagent Delivery
  • The reagent delivery system of the invention can be any system that allows the delivery of reagents to discrete portions of the array in order to keep the integrity of the defined spatial patterns of the encoding scheme. Such discrete delivery can be achieved in a number of different ways.
  • In one exemplary aspect, the reagent delivery system can be a flow-based system. The flow-based systems for reagent delivery in the present invention can include one or more pumps, valves, fluid reservoirs, channels, and/or reagent storage cells. Such a reagent delivery system is configured to move fluid in contact with a discrete section of the substrate surface. Movement of the reagents can be driven through a fluid by a pump disposed, for example, downstream of the fluid reagents. The pump can drive each fluid reagent to (and past) the reaction compartment. Alternatively, the reagents may be driven through the fluid by gravity.
  • US Appln Nos. 20070166725 and 20050239192 disclose certain general- purpose fluidics tools that can be used with the assay systems of the invention. These allow the precise manipulation of gases, liquids and solids to accomplish very complex analytical manipulations with relatively simple hardware.
  • In a more specific example, one or more flow-cells can be attached to the substrate from above. The flow-cell can include inlet and outlet tubes connected thereto and optionally an external pump can be used to deliver the sample or reagents to the flow-cell and across the substrate. The flow cell is configured to deliver reagents only to certain portions of the array, restricting the amount and type of reagent delivered to any specific section of the array.
  • In another aspect, a microfluidic system can be integrated into the substrate or externally attached on top of the substrate. Microfluidic passages for holding and carrying fluid can be formed on and/or above the planar substrate by a fluidics layer abutted to the substrate. Fluid reagents can be selected according to selective opening and closing of valves disposed between reagent reservoirs.
  • Pumps generally include any mechanism for moving fluid and/or reagents disposed in fluid. In some examples, the pump can be configured to move fluid and/or reagents through passages with small volumes (i.e., microfluidic structures). The pump can operate mechanically by exerting a positive or negative pressure on fluid and/or on a structure carrying fluid, electrically by appropriate application of an electric field(s), or both, among others. Exemplary mechanical pumps may include syringe pumps, peristaltic pumps, rotary pumps, pressurized gas, pipettors, etc. The mechanical pumps may be micromachined, molded, etc. Exemplary electrical pumps can include electrodes and may operate by electrophoresis, electroendoosmosis, electrocapillarity, dielectrophoresis (including traveling wave forms thereof), and/or the like.
  • Valves generally include any mechanism for regulating the passage of fluid through a channel. The valves can include, for example, deformable members that can be selectively deformed to partially or completely close a channel, a movable projection that can be selectively extended into the channel to partially or completely block the channel, an electrocapillary structure, and/or the like.
  • In yet another aspect, an open gasket can be attached to the top of the substrate and the sample and reagents can be injected into the gasket. Suitable gasket materials include, but are not limited to, neoprene, nitrile, and silicone rubber. Alternatively, a watertight reaction chamber formed by a gasket sandwiched between the substrate and a chemically inert, water resistant material such as, but not limited to, black-anodized aluminum, thermoplastics (e.g., polystyrene, polycarbonate, etc), glass, etc.
  • In a specific aspect of the present invention, the delivery system can comprise a microcircuit arrangement including an imager, such as a CCD or IGFET-based (e.g., CMOS-based) imager and an ultrasonic sprayer for reagent delivery such as described in US Appln No. 20090197326, which is incorporated herein by reference.
  • In yet another aspect of the invention, the reagent delivery system controls the delivery of reagents to specific patterns on a substrate surface using semiconductor techniques such as masking and spraying. Specific areas of a substrate surface can be protected from exposure to reagents through use of a mask to protect specific areas from exposure. The reagents may be introduced to the substrate using conventional techniques such as spraying or fluid flow. The use of the masked substrate delivery results in a patterned delivery scheme on the substrate surface.
  • In a preferred aspect of the invention, the reagent delivery instrumentation is based on inkjet printing technology. There are a variety of different ink-jetting mechanisms (e.g., thermal, piezoelectric) and compatibility has been shown with aqueous and organic ink formulations. Sets of independently actuated nozzles can be used to deliver multiple reagents at the same time, and very high resolutions can be achieved.
  • Software for use in the Assay System
  • In order to target specific sites of interest, an informative image of the biological section to be analyzed can be used to assist in the reagent delivery methods and associated encoding scheme. Sample regions can be identified using image processing (e.g., images of cell types differentiated by immunohistochemistry or other staining chemistries) integrated with the other features of the assay system. In some aspects, software is used to automatically translate this information into a reagent delivery pattern. A mechanism to register and align very precisely the biological sample in a targeting system is thus a preferred component of the assay systems of the invention. Mechanisms such as the use of fiducial markers on slides and other very accurate physical positioning systems can be adapted to this purpose.
  • Additional software components will also be key components that will be part of a complete assay system. The invention thus preferably comprises a complete suite of software tailored to the assay system. Optionally, oligonucleotide design software will be customized for the specific assay to be run, and may be integrated as a part of the system. Also optionally, algorithms and software for data analysis may be integrated to assist in determination of results of the assays. This can be especially useful, as the type of dataset that will be generated will be novel, particularly as a consequence of scale. The ability to provide algorithms and software tools that are specifically designed for analysis of spatially-associated data for significant patterns, including pattern-analysis software and visualization tools, is a novel feature that will enhance the value of the data generated by the assay systems.
  • In certain aspects, the assay system will comprise processes for making and carrying out quality control of reagents, e.g., the integrity and sequence fidelity of oligonucleotide pools. In particular, reagents will need to be formulated for compatibility with the reagent delivery instrumentation. Factors such as volatility, stability at key temperatures, and chemical compatibility can be optimized by those skilled in the art upon reading the present disclosure.
  • Applications of Assay System
  • It will be apparent to one skilled in the art upon reading the present disclosure that there are numerous very important areas of biological research, diagnostics, and drug development that will benefit from a high throughput means to simultaneously measure the presence or absence and spatial location of a biological molecule in a sample. For example, this technology combining the ability to analyze semi-quantitatively the expression of many different genes with the ability to image the spatial organization of expression across many cells in a tissue is enabling for many different areas of basic research. The following are exemplary uses and are by no means meant to be limiting in scope.
  • In one example, 3-dimensional patterns of expression can be determined by analyzing a series of tissue sections, in a manner analogous to image reconstruction in CT scanning. This can be used to measure changes in gene expression in disease pathology, e.g., in cancerous tissue and/or a tissue upon injury, inflammation or infection. With the assay systems of the invention, more detailed information on gene expression and protein localization in complex tissues can be obtained. This may lead to new insights into the function and regulation both in normal and diseased states, and is likely to provide new hypotheses that can be tested. For example, a system of the invention may enable some of the insights gained from many individual studies and larger programs like ENCODE (Birney et al., 2007) and modENCODE to be integrated at the tissue level. The assay systems will also aid in computational efforts to model interacting networks of gene expression in the field of systems biology.
  • The assay systems also provide a novel approach that enables the analysis of somatic variation, e.g., somatic mutations in cancer or variability in response to infectious organisms. For example, tumors are typically highly heterogeneous, containing cancer cells as well as genetically normal cells in an abnormal local environment. Cancer cells undergo mutation and selection, and in this process it is not unusual for local clones to develop. Identifying relatively rare somatic mutations in the context of tumors may enable the study of the role of key mutations in the selection of clonal variants. Transcriptional patterns associated with angiogenesis, inflammation, or other cancer related processes in both cancer and genetically normal cells can be analyzed for insights into cancer biology and assist in the development of new therapeutic agents for the treatment of cancers.
  • In another example, different people have varying susceptibility to infectious organisms, and much of this may be to underlying genetic differences in individuals and/or populations. Identifying these differences will aid in an understanding of the underlying disease pathologies and assist in the development of vaccines or therapeutics to prevent or ameliorate these disease states.
  • Importantly, in addition to providing spatially associated information, the technology of the invention will allow a great increase in the sensitivity of detecting rare mutations. The reason is that signal to noise can be dramatically increased because the approach of the invention assays a small location in any given reaction. In a typical assay for rare mutations in a mixed sample, the sample is treated in bulk, i.e. nucleic acids are extracted from many cells into a single pool. Thus, if a mutation is present in 1 cell in 10,000, it must be detected against a background of normal DNA from 10,000 cells. In contrast, with the systems of the invention many cells can be analyzed, but individual cells or small groups of cells would be identified by the spatial coding system. Therefore, the background can be reduced by orders of magnitude, greatly increasing sensitivity. Furthermore, the spatial organization of mutant cells can be observed. This may be particularly important in detecting key mutations in tissue sections in cancer. Already, molecular histological analyses are yielding insights into cancer biology and may have potential for use in diagnostics (Choe et al., 2003). The technology of the invention promises to greatly increase the power of such approaches.
  • EXAMPLES
  • The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventor regards as his invention, nor are they intended to represent or imply that the experiments below are all of or the only experiments performed. It will be appreciated by persons skilled in the art that numerous variations and/or modifications may be made to the invention as shown in the specific embodiments without departing from the spirit or scope of the invention as broadly described. The present embodiments are, therefore, to be considered in all respects as illustrative and not restrictive.
  • Efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees centigrade, and pressure is at or near atmospheric.
  • Example 1: Initial Proof of Concept of Encoding Scheme
  • As an initial proof of concept, a model system is developed using a microarray to demonstrate a working single-plex assay. The basic design validates the concept of the assay, and establishes a working assay prior to addressing issues related to the analysis of a more complicated biological sample. Conventional sequencing is used as a readout for this proof of concept.
  • A microarray is used as a proxy for a tissue section. The target sequences of the microarray are fully specified, so that the composition of the targets are known and can be varied systematically. Synthetic oligonucleotide templates are attached to a glass slide via a 5′ amino modification. Each slide has a single oligonucleotide template sequence, and the assays that are carried out may employ either ligation, or extension followed by ligation as this may be useful in determining certain polymorphisms.
  • Once the in situ part of the assay is complete, the reaction products are eluted and analyzed by qPCR to determined presence or absence of a product and estimate yield, and by conventional sequencing to determine the structure of the assay products. The single plex assays that are tested include appropriate positive and negative controls, and a single nucleotide variant (SNV) to check ability to discriminate single base variants.
  • Example 2: Scalability
  • The complexity of the assay system is increased to establish scalability of the assay for use in high throughput studies. Scalability of both the spatial encoding and assay systems is demonstrated by carrying out a 24-plex×24-site assay using a microarray model system.
  • The amount of biological target, here a DNA target sequence, at each assay location is systematically varied on microarray substrate. For example, in a microarray with 50 micron spot size (center to center), a 1 mm2 area contains ˜400 spots. The region around each site is optionally occupied by a region that is devoid of these spots to allow individual resolvability of the target sequences. Alternatively, the spots may be clustered, with two or more directly adjacent spots surrounded by or adjacent to a region that is devoid of target sequences.
  • In order to demonstrate that spatial encoding is accurate, the sites comprise different target compositions to show that the assay readout matches the expected composition of each site. With 24 target sequences, a simple digital pattern is made with each site having a different set of 12 targets present and 12 targets absent, to make a binary code (0=absent, 1=present). The assay readout is then determined to show that the detected regions match the expected signal after spatial decoding. In this particular example, the code space is large enough (224) so that even a few errors would not result in different codes being mixed up. Moreover, this design allows identification of errors and allows an estimation not only of accuracy of spatial encoding but also of accuracy calling the presence or absence of target sequences. In an exemplary aspect, a 4×4 arrangement of 16 sequences is used for the array configuration. A white square indicates that the sequence is absent and a black square that it is present, i.e. 8 of the 16 possible sequences are present in this sample. In a different sample, a different pattern of absent and present sequences can be constructed. In this way, unique patterns are associated with spatial locations so that the accuracy of spatial encoding can be measured.
  • The ability to detect quantitative differences is evaluated by generating dose-response curves for each of the 24 assays that are carried out at each site in a 24-site assay. This allows estimation of the limit of detection, dynamic range, and power to detect a given fold-change across the range.
  • In one aspect, a latin square design is used to represent individual targets at different ratios by varying the number of features for each target. In other words, with multiple spots in a site, the number of spots allocated to each of the 24 target sequences can be varied and each of the 24 sites can have a different composition. A 1×3 inch microarray is sufficiently large to permit multiple replicates. This larger set of 24 sequences will require deconvolution, and this is accomplished using high throughput techniques such as next-generation sequencing technologies (e.g., SOLiD™ technology (Life Technologies, Inc., Carlsbad, Calif.) or Genome Analyzer (Illumina, Inc., San Diego, Calif.)). The use of the 24-plex assay demonstrates both the accuracy of spatial encoding and decoding, and the quantitative response of the assay system.
  • Example 3: Adaptation of the Assay to Preserved Samples
  • Genomic DNA is assayed as a proof of concept for assaying RNA, as it provides a way to establish a single-copy reference signal. Once a working assay is developed for FFPE samples, it is adapted to an RNA assay. To this end, assay oligonucleotide concentrations are assayed to ensure compatibility with high multiplexing. Assuming a cell diameter of 10 microns, and delivery of a 10 micron diameter reagent droplet to an individual cell, the volume of the droplet will be ˜500 μl and can contain ˜3×1011 molecules at a 1 μM concentration. To assay 1,000 target sequences in 10,000 cells, 2,000 targeting oligonucleotides would be required in a droplet. Therefore, each droplet could contain ˜160 million copies of each assay oligo, a vast excess over the few thousand target sequences in a cell.
  • The handling of small absolute numbers of product molecules generated from very small or compromised samples are enhanced to counter the issue of low recovery efficiency; that is, elution is efficient and losses resulting from adsorption of molecules to surfaces are prevented. An approach to addressing the latter issue is to include a carrier material, such as glycogen or carrier nucleic acids.
  • Example 4: Adapting the Assay to a Biological Sample
  • A control RNA template is immobilized to a solid support in order to create an artificial system. The assay is performed using T4 DNA ligase, which can repair nicks in DNA/RNA hybrids. Assays are carried out on matched slides, or different sections of the same slide, where in one case gDNA is assayed and in the other RNA is assayed. When assaying gDNA the slide can be pretreated with RNase, and when assaying RNA the slide is pretreated with DNase. Results of the assay are confirmed by extracting gDNA or RNA and quantitating the relative amounts by qPCR or RT-qPCR respectively.
  • In order make the tissue section RNA assays as informative as possible, pre-existing information on expression levels in specific tissues to target transcripts across a range of abundances are used in the assay design. Both high abundance transcripts, as well as some medium and low abundance transcripts, are targeted to enable an initial assessment of the quantitative performance characteristics of the assay.
  • The preceding merely illustrates the principles of the invention. It will be appreciated that those skilled in the art will be able to devise various arrangements which, although not explicitly described or shown herein, embody the principles of the invention and are included within its spirit and scope. Furthermore, all examples and conditional language recited herein are principally intended to aid the reader in understanding the principles of the invention and the concepts contributed by the inventors to furthering the art, and are to be construed as being without limitation to such specifically recited examples and conditions. Moreover, all statements herein reciting principles, aspects, and embodiments of the invention as well as specific examples thereof, are intended to encompass both structural and functional equivalents thereof. Additionally, it is intended that such equivalents include both currently known equivalents and equivalents developed in the future, i.e., any elements developed that perform the same function, regardless of structure. The scope of the present invention, therefore, is not intended to be limited to the exemplary embodiments shown and described herein. Rather, the scope and spirit of present invention is embodied by the appended claims. In the claims that follow, unless the term “means” is used, none of the features or elements recited therein should be construed as means-plus-function limitations pursuant to 35 U.S.C. § 112, ¶16.

Claims (30)

1. (canceled)
2. A system for analyzing a target biological molecule of a tissue sample, the system comprising:
a laser configured to deliver radiation to a tissue sample comprising a plurality of probes, wherein a probe of the plurality of probes comprises a peptide that specifically binds a target biological molecule of the tissue sample and a mass tag conjugated to the peptide;
a matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) detection apparatus comprising a mass analyzer configured to:
receive a chemical moiety comprising at least a portion of the mass tag after liberation of the chemical moiety from a location of interest in the sample; and
detect and mass analyze the at least a portion of the mass tag.
3. The system of claim 2, wherein the detection apparatus is configured to determine a relative abundance of the at least a portion of the mass tag from the location of interest in the tissue sample.
4. The system of claim 2, wherein the laser is configured to liberate the chemical moiety from the location of interest in the tissue sample.
5. The system of claim 2, wherein the chemical moiety is the mass tag.
6. The system of claim 2, wherein the peptide specifically binds a complementary region of the target biological molecule.
7. The system of claim 2, wherein the probe is a first probe comprising a first peptide that specifically binds a first target biological molecule of the tissue sample and a first mass tag conjugated to the first peptide, and wherein a second probe of the plurality of probes comprises a second peptide that specifically binds a second target biological molecule of the tissue sample and a second mass tag conjugated to the second peptide.
8. The system of claim 7, wherein the first and second mass tags are different.
9. The system of claim 7, wherein the first and second target biological molecules are the same.
10. The system of claim 7, wherein the first and second target biological molecules are different.
11. The system of claim 2, further comprising a processing circuit comprising instructions that, when executed, cause the processing circuit to obtain information about the location of interest in the tissue sample.
12. The system of claim 11, wherein the processing circuit is configured to obtain information about multiple locations of interest in the tissue sample, and the detection apparatus is configured to determine information corresponding to a relative abundance of the at least a portion of the mass tag at different locations in the tissue sample corresponding to the multiple locations of interest.
13. The system of claim 11, wherein the processing circuit comprises instructions that, when executed, cause the processing circuit to receive the information about the location of interest in the tissue sample from a user of the system.
14. The system of claim 2, wherein the target biological molecule comprises a protein.
15. The system of claim 2, wherein the tissue sample comprises a fresh-frozen or formalin-fixed paraffin embedded (FFPE) tissue sample.
16. The system of claim 2, wherein the peptide forms a portion of an antibody that specifically binds the target biological molecule.
17. The system of claim 12, wherein each of the multiple locations of interest corresponds to a different group of one or more cells in the tissue sample.
18. The system of claim 12, wherein each of the multiple locations of interest corresponds to a different cell type in the tissue sample.
19. The system of claim 12, wherein each of the multiple locations of interest corresponds to a different cell in the tissue sample.
20. A method for analyzing a target biological molecule of a tissue sample, the method comprising:
obtaining a tissue sample comprising a plurality of probes, wherein a probe of the plurality of probes comprises a peptide that specifically binds a target biological molecule of the tissue sample and a mass tag conjugated to the peptide;
directing radiation to the tissue sample to liberate a chemical moiety comprising at least a portion of the mass tag from a location of interest in the sample; and
mass analyzing the at least a portion of the mass tag.
21. The method of claim 20, further comprising determining a relative abundance of the at least a portion of the mass tag at the location of interest in the tissue sample.
22. The method of claim 20, comprising liberating the chemical moiety from the location of interest in the tissue by ionizing the chemical moiety.
23. The method of claim 22, wherein the chemical moiety is the mass tag.
24. The method of claim 20, wherein the peptide of the probe specifically binds a complementary region of the target biological molecule.
25. The method of claim 20, wherein the probe is a first probe comprising a first peptide that specifically binds a first target biological molecule of the tissue sample and a first mass tag conjugated to the first peptide, and wherein a second probe of the plurality of probes comprises a second peptide that specifically binds a second target biological molecule of the tissue sample and a second mass tag conjugated to the second peptide.
26. The method of claim 25, wherein the first and second mass tags are different.
27. The method of claim 20, further comprising determining information corresponding to a relative abundance of the at least a portion of the mass tag at different locations in the tissue sample corresponding to multiple locations of interest in the tissue sample.
28. The method of claim 20, wherein the target biological molecule comprises a protein.
29. The method of claim 20, wherein the peptide forms a portion of an antibody that specifically binds the target biological molecule.
30. The method of claim 27, wherein each of the multiple locations of interest corresponds to one of: a different group of one or more cells in the tissue sample, and a different cell type in the tissue sample.
US16/437,726 2010-04-05 2019-06-11 Spatially Encoded Biological Assays Abandoned US20190300945A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US16/437,726 US20190300945A1 (en) 2010-04-05 2019-06-11 Spatially Encoded Biological Assays
US16/443,771 US20190309355A1 (en) 2010-04-05 2019-06-17 Spatially Encoded Biological Assays
US16/660,234 US10662468B2 (en) 2010-04-05 2019-10-22 Spatially encoded biological assays
US16/670,603 US10612079B2 (en) 2010-04-05 2019-10-31 Spatially encoded biological assays
US16/734,216 US10619196B1 (en) 2010-04-05 2020-01-03 Spatially encoded biological assays
US16/837,924 US10983113B2 (en) 2010-04-05 2020-04-01 Spatially encoded biological assays
US16/913,672 US11733238B2 (en) 2010-04-05 2020-06-26 Spatially encoded biological assays
US18/341,373 US20230358733A1 (en) 2010-04-05 2023-06-26 Spatially Encoded Biological Assays

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US32112410P 2010-04-05 2010-04-05
US13/080,616 US9371598B2 (en) 2010-04-05 2011-04-05 Spatially encoded biological assays
US15/187,661 US10308982B2 (en) 2010-04-05 2016-06-20 Spatially encoded biological assays
US16/276,235 US10480022B2 (en) 2010-04-05 2019-02-14 Spatially encoded biological assays
US16/402,098 US10472669B2 (en) 2010-04-05 2019-05-02 Spatially encoded biological assays
US16/414,213 US10787701B2 (en) 2010-04-05 2019-05-16 Spatially encoded biological assays
US16/430,015 US20190300943A1 (en) 2010-04-05 2019-06-03 Spatially Encoded Biological Assays
US16/437,726 US20190300945A1 (en) 2010-04-05 2019-06-11 Spatially Encoded Biological Assays

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/430,015 Continuation US20190300943A1 (en) 2010-04-05 2019-06-03 Spatially Encoded Biological Assays

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/443,771 Continuation US20190309355A1 (en) 2010-04-05 2019-06-17 Spatially Encoded Biological Assays
US16/913,672 Continuation US11733238B2 (en) 2010-04-05 2020-06-26 Spatially encoded biological assays

Publications (1)

Publication Number Publication Date
US20190300945A1 true US20190300945A1 (en) 2019-10-03

Family

ID=68055160

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/437,726 Abandoned US20190300945A1 (en) 2010-04-05 2019-06-11 Spatially Encoded Biological Assays
US16/913,672 Active US11733238B2 (en) 2010-04-05 2020-06-26 Spatially encoded biological assays

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/913,672 Active US11733238B2 (en) 2010-04-05 2020-06-26 Spatially encoded biological assays

Country Status (1)

Country Link
US (2) US20190300945A1 (en)

Cited By (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11162132B2 (en) 2015-04-10 2021-11-02 Spatial Transcriptomics Ab Spatially distinguished, multiplex nucleic acid analysis of biological specimens
US11286515B2 (en) 2013-06-25 2022-03-29 Prognosys Biosciences, Inc. Methods and systems for determining spatial patterns of biological targets in a sample
US11332790B2 (en) 2019-12-23 2022-05-17 10X Genomics, Inc. Methods for spatial analysis using RNA-templated ligation
US11352659B2 (en) 2011-04-13 2022-06-07 Spatial Transcriptomics Ab Methods of detecting analytes
US11407992B2 (en) 2020-06-08 2022-08-09 10X Genomics, Inc. Methods of determining a surgical margin and methods of use thereof
US11408029B2 (en) 2020-06-25 2022-08-09 10X Genomics, Inc. Spatial analysis of DNA methylation
US11434524B2 (en) 2020-06-10 2022-09-06 10X Genomics, Inc. Methods for determining a location of an analyte in a biological sample
US11512308B2 (en) 2020-06-02 2022-11-29 10X Genomics, Inc. Nucleic acid library methods
US11519033B2 (en) 2018-08-28 2022-12-06 10X Genomics, Inc. Method for transposase-mediated spatial tagging and analyzing genomic DNA in a biological sample
US11535887B2 (en) 2020-04-22 2022-12-27 10X Genomics, Inc. Methods for spatial analysis using targeted RNA depletion
US11560592B2 (en) 2020-05-26 2023-01-24 10X Genomics, Inc. Method for resetting an array
US11592447B2 (en) 2019-11-08 2023-02-28 10X Genomics, Inc. Spatially-tagged analyte capture agents for analyte multiplexing
US11608520B2 (en) 2020-05-22 2023-03-21 10X Genomics, Inc. Spatial analysis to detect sequence variants
US11618897B2 (en) 2020-12-21 2023-04-04 10X Genomics, Inc. Methods, compositions, and systems for capturing probes and/or barcodes
US11624086B2 (en) 2020-05-22 2023-04-11 10X Genomics, Inc. Simultaneous spatio-temporal measurement of gene expression and cellular activity
US11649485B2 (en) 2019-01-06 2023-05-16 10X Genomics, Inc. Generating capture probes for spatial analysis
US11692218B2 (en) 2020-06-02 2023-07-04 10X Genomics, Inc. Spatial transcriptomics for antigen-receptors
US11702693B2 (en) 2020-01-21 2023-07-18 10X Genomics, Inc. Methods for printing cells and generating arrays of barcoded cells
US11702698B2 (en) 2019-11-08 2023-07-18 10X Genomics, Inc. Enhancing specificity of analyte binding
US11732299B2 (en) 2020-01-21 2023-08-22 10X Genomics, Inc. Spatial assays with perturbed cells
US11732300B2 (en) 2020-02-05 2023-08-22 10X Genomics, Inc. Increasing efficiency of spatial analysis in a biological sample
US11739381B2 (en) 2021-03-18 2023-08-29 10X Genomics, Inc. Multiplex capture of gene and protein expression from a biological sample
US11753673B2 (en) 2021-09-01 2023-09-12 10X Genomics, Inc. Methods, compositions, and kits for blocking a capture probe on a spatial array
US11761038B1 (en) 2020-07-06 2023-09-19 10X Genomics, Inc. Methods for identifying a location of an RNA in a biological sample
US11768175B1 (en) 2020-03-04 2023-09-26 10X Genomics, Inc. Electrophoretic methods for spatial analysis
US11821035B1 (en) 2020-01-29 2023-11-21 10X Genomics, Inc. Compositions and methods of making gene expression libraries
US11827935B1 (en) 2020-11-19 2023-11-28 10X Genomics, Inc. Methods for spatial analysis using rolling circle amplification and detection probes
US11835462B2 (en) 2020-02-11 2023-12-05 10X Genomics, Inc. Methods and compositions for partitioning a biological sample
US11891654B2 (en) 2020-02-24 2024-02-06 10X Genomics, Inc. Methods of making gene expression libraries
US11898205B2 (en) 2020-02-03 2024-02-13 10X Genomics, Inc. Increasing capture efficiency of spatial assays
US11926863B1 (en) 2020-02-27 2024-03-12 10X Genomics, Inc. Solid state single cell method for analyzing fixed biological cells
US11926867B2 (en) 2019-01-06 2024-03-12 10X Genomics, Inc. Generating capture probes for spatial analysis
US11926822B1 (en) 2020-09-23 2024-03-12 10X Genomics, Inc. Three-dimensional spatial analysis
US11933957B1 (en) 2018-12-10 2024-03-19 10X Genomics, Inc. Imaging system hardware
US11965213B2 (en) 2019-05-30 2024-04-23 10X Genomics, Inc. Methods of detecting spatial heterogeneity of a biological sample
US11970739B2 (en) 2023-07-06 2024-04-30 10X Genomics, Inc. Multiplex capture of gene and protein expression from a biological sample

Family Cites Families (750)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4514388A (en) 1983-03-22 1985-04-30 Psaledakis Nicholas G Proteolytic enzymes in the zymogen form to treat sarcoma cells
US4574729A (en) 1984-08-06 1986-03-11 E. I. Du Pont De Nemours & Co. Chamber block for a cytocentrifuge having centrifugal force responsive supernatant withdrawal means
US5061049A (en) 1984-08-31 1991-10-29 Texas Instruments Incorporated Spatial light modulator and method
US4965188A (en) 1986-08-22 1990-10-23 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences using a thermostable enzyme
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4883867A (en) 1985-11-01 1989-11-28 Becton, Dickinson And Company Detection of reticulocytes, RNA or DNA
US4800159A (en) 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
US5589173A (en) 1986-11-04 1996-12-31 Genentech, Inc. Method and therapeutic compositions for the treatment of myocardial infarction
US5525464A (en) 1987-04-01 1996-06-11 Hyseq, Inc. Method of sequencing by hybridization of oligonucleotide probes
US4968601A (en) 1988-02-09 1990-11-06 The United States Of America As Represented By The Dept. Of Health & Human Services Method for diagnosing latent viral infection
GB8810400D0 (en) 1988-05-03 1988-06-08 Southern E Analysing polynucleotide sequences
US4988617A (en) 1988-03-25 1991-01-29 California Institute Of Technology Method of detecting a nucleotide change in nucleic acids
US5130238A (en) 1988-06-24 1992-07-14 Cangene Corporation Enhanced nucleic acid amplification process
AU2684488A (en) 1988-06-27 1990-01-04 Carter-Wallace, Inc. Test device and method for colored particle immunoassay
US5512439A (en) 1988-11-21 1996-04-30 Dynal As Oligonucleotide-linked magnetic particles and uses thereof
US5002882A (en) 1989-04-27 1991-03-26 New England Biolabs, Inc. Method for producing the XmaI restriction endonuclease and methylase
RU2145635C1 (en) 1989-05-18 2000-02-20 Чирон Корпорейшн Oligomer (variants), method of detection of hcv sequence (variants), set for detection, method of blood preparing
WO1991006678A1 (en) 1989-10-26 1991-05-16 Sri International Dna sequencing
US5494810A (en) 1990-05-03 1996-02-27 Cornell Research Foundation, Inc. Thermostable ligase-mediated DNA amplifications system for the detection of genetic disease
US5559032A (en) 1990-06-29 1996-09-24 Pomeroy; Patrick C. Method and apparatus for post-transfer assaying of material on solid support
US5455166A (en) 1991-01-31 1995-10-03 Becton, Dickinson And Company Strand displacement amplification
US5183053A (en) 1991-04-12 1993-02-02 Acuderm, Inc. Elliptical biopsy punch
WO1993004199A2 (en) 1991-08-20 1993-03-04 Scientific Generics Limited Methods of detecting or quantitating nucleic acids and of producing labelled immobilised nucleic acids
US5474796A (en) 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
US6872816B1 (en) 1996-01-24 2005-03-29 Third Wave Technologies, Inc. Nucleic acid detection kits
US6759226B1 (en) 2000-05-24 2004-07-06 Third Wave Technologies, Inc. Enzymes for the detection of specific nucleic acid sequences
CA2119126C (en) 1991-09-16 1996-09-03 Stephen T. Yue Dimers of unsymmetrical cyanine dyes
US5321130A (en) 1992-02-10 1994-06-14 Molecular Probes, Inc. Unsymmetrical cyanine dyes with a cationic side chain
US5308751A (en) 1992-03-23 1994-05-03 General Atomics Method for sequencing double-stranded DNA
US5503980A (en) 1992-11-06 1996-04-02 Trustees Of Boston University Positional sequencing by hybridization
US5472881A (en) 1992-11-12 1995-12-05 University Of Utah Research Foundation Thiol labeling of DNA for attachment to gold surfaces
US5410030A (en) 1993-04-05 1995-04-25 Molecular Probes, Inc. Dimers of unsymmetrical cyanine dyes containing pyridinium moieties
US5436134A (en) 1993-04-13 1995-07-25 Molecular Probes, Inc. Cyclic-substituted unsymmetrical cyanine dyes
US5658751A (en) 1993-04-13 1997-08-19 Molecular Probes, Inc. Substituted unsymmetrical cyanine dyes with selected permeability
US5837832A (en) 1993-06-25 1998-11-17 Affymetrix, Inc. Arrays of nucleic acid probes on biological chips
US5874239A (en) 1993-07-30 1999-02-23 Affymax Technologies N.V. Biotinylation of proteins
US6401267B1 (en) 1993-09-27 2002-06-11 Radoje Drmanac Methods and compositions for efficient nucleic acid sequencing
US5610287A (en) 1993-12-06 1997-03-11 Molecular Tool, Inc. Method for immobilizing nucleic acid molecules
SE9400522D0 (en) 1994-02-16 1994-02-16 Ulf Landegren Method and reagent for detecting specific nucleotide sequences
US5512462A (en) 1994-02-25 1996-04-30 Hoffmann-La Roche Inc. Methods and reagents for the polymerase chain reaction amplification of long DNA sequences
US5677170A (en) 1994-03-02 1997-10-14 The Johns Hopkins University In vitro transposition of artificial transposons
JPH09510351A (en) 1994-03-16 1997-10-21 ジェン−プローブ・インコーポレイテッド Isothermal strand displacement nucleic acid amplification method
US6015880A (en) 1994-03-16 2000-01-18 California Institute Of Technology Method and substrate for performing multiple sequential reactions on a matrix
US5552278A (en) 1994-04-04 1996-09-03 Spectragen, Inc. DNA sequencing by stepwise ligation and cleavage
US5807522A (en) 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US5641658A (en) 1994-08-03 1997-06-24 Mosaic Technologies, Inc. Method for performing amplification of nucleic acid with two primers bound to a single solid support
EP0777749B1 (en) 1994-08-19 2002-10-30 PE Corporation (NY) Coupled amplification and ligation method
US5846719A (en) 1994-10-13 1998-12-08 Lynx Therapeutics, Inc. Oligonucleotide tags for sorting and identification
KR960022566A (en) 1994-12-30 1996-07-18 김충환 Novel aminooligosaccharide derivatives and preparation method thereof
US5736351A (en) 1995-01-09 1998-04-07 New Horizons Diagnostics Corporation Method for detection of contaminants
US5959098A (en) 1996-04-17 1999-09-28 Affymetrix, Inc. Substrate preparation process
US5750341A (en) 1995-04-17 1998-05-12 Lynx Therapeutics, Inc. DNA sequencing by parallel oligonucleotide extensions
US5648245A (en) 1995-05-09 1997-07-15 Carnegie Institution Of Washington Method for constructing an oligonucleotide concatamer library by rolling circle replication
US5928928A (en) 1995-06-07 1999-07-27 Universiteit Van Amsterdam Human chitinase, its recombinant production, its use for decomposing chitin, its use in therapy or prophylaxis against infection diseases
SE504798C2 (en) 1995-06-16 1997-04-28 Ulf Landegren Immunoassay and test kits with two reagents that can be cross-linked if adsorbed to the analyte
WO1997013845A2 (en) 1995-10-13 1997-04-17 President And Fellows Of Harvard College Phosphopantetheinyl transferases and uses thereof
US5716825A (en) 1995-11-01 1998-02-10 Hewlett Packard Company Integrated nucleic acid analysis system for MALDI-TOF MS
US5763175A (en) 1995-11-17 1998-06-09 Lynx Therapeutics, Inc. Simultaneous sequencing of tagged polynucleotides
US5854033A (en) 1995-11-21 1998-12-29 Yale University Rolling circle replication reporter systems
US6300063B1 (en) 1995-11-29 2001-10-09 Affymetrix, Inc. Polymorphism detection
EP0880598A4 (en) 1996-01-23 2005-02-23 Affymetrix Inc Nucleic acid analysis techniques
US6913881B1 (en) 1996-01-24 2005-07-05 Third Wave Technologies, Inc. Methods and compositions for detecting target sequences
US5985557A (en) 1996-01-24 1999-11-16 Third Wave Technologies, Inc. Invasive cleavage of nucleic acids
US6875572B2 (en) 1996-01-24 2005-04-05 Third Wave Technologies, Inc. Nucleic acid detection assays
AU735440B2 (en) 1996-02-09 2001-07-05 Cornell Research Foundation Inc. Detection of nucleic acid sequence differences using the ligase detection reaction with addressable arrays
US6852487B1 (en) 1996-02-09 2005-02-08 Cornell Research Foundation, Inc. Detection of nucleic acid sequence differences using the ligase detection reaction with addressable arrays
US6013440A (en) 1996-03-11 2000-01-11 Affymetrix, Inc. Nucleic acid affinity columns
US5928906A (en) 1996-05-09 1999-07-27 Sequenom, Inc. Process for direct sequencing during template amplification
EP0912761A4 (en) 1996-05-29 2004-06-09 Cornell Res Foundation Inc Detection of nucleic acid sequence differences using coupled ligase detection and polymerase chain reactions
US20050003431A1 (en) 1996-08-16 2005-01-06 Wucherpfennig Kai W. Monovalent, multivalent, and multimeric MHC binding domain fusion proteins and conjugates, and uses therefor
US5925545A (en) 1996-09-09 1999-07-20 Wisconsin Alumni Research Foundation System for in vitro transposition
US5965443A (en) 1996-09-09 1999-10-12 Wisconsin Alumni Research Foundation System for in vitro transposition
US6221654B1 (en) 1996-09-25 2001-04-24 California Institute Of Technology Method and apparatus for analysis and sorting of polynucleotides based on size
DE19639673A1 (en) 1996-09-27 1998-04-09 Daimler Benz Ag Display arranged in a motor vehicle in the region of the windscreen
GB9620209D0 (en) 1996-09-27 1996-11-13 Cemu Bioteknik Ab Method of sequencing DNA
US6083761A (en) 1996-12-02 2000-07-04 Glaxo Wellcome Inc. Method and apparatus for transferring and combining reagents
US6060240A (en) 1996-12-13 2000-05-09 Arcaris, Inc. Methods for measuring relative amounts of nucleic acids in a complex mixture and retrieval of specific sequences therefrom
US5837466A (en) 1996-12-16 1998-11-17 Vysis, Inc. Devices and methods for detecting nucleic acid analytes in samples
GB9626815D0 (en) 1996-12-23 1997-02-12 Cemu Bioteknik Ab Method of sequencing DNA
US6737236B1 (en) 1997-01-08 2004-05-18 Proligo, Llc Bioconjugation of macromolecules
US6309824B1 (en) 1997-01-16 2001-10-30 Hyseq, Inc. Methods for analyzing a target nucleic acid using immobilized heterogeneous mixtures of oligonucleotide probes
DK0971946T3 (en) 1997-01-21 2006-10-30 Gen Hospital Corp Selection of proteins using RNA-protein fusions
US6261804B1 (en) 1997-01-21 2001-07-17 The General Hospital Corporation Selection of proteins using RNA-protein fusions
US8207093B2 (en) 1997-01-21 2012-06-26 The General Hospital Corporation Selection of proteins using RNA-protein fusions
US5837860A (en) 1997-03-05 1998-11-17 Molecular Tool, Inc. Covalent attachment of nucleic acid molecules onto solid-phases via disulfide bonds
US6327410B1 (en) 1997-03-14 2001-12-04 The Trustees Of Tufts College Target analyte sensors utilizing Microspheres
US7622294B2 (en) 1997-03-14 2009-11-24 Trustees Of Tufts College Methods for detecting target analytes and enzymatic reactions
US6023540A (en) 1997-03-14 2000-02-08 Trustees Of Tufts College Fiber optic sensor with encoded microspheres
KR20010005544A (en) 1997-03-21 2001-01-15 그레그 펄쓰 Extraction and utilisation of VNTR alleles
AU6846798A (en) 1997-04-01 1998-10-22 Glaxo Group Limited Method of nucleic acid sequencing
EP3034626A1 (en) 1997-04-01 2016-06-22 Illumina Cambridge Limited Method of nucleic acid sequencing
US6143496A (en) 1997-04-17 2000-11-07 Cytonix Corporation Method of sampling, amplifying and quantifying segment of nucleic acid, polymerase chain reaction assembly having nanoliter-sized sample chambers, and method of filling assembly
US6969488B2 (en) 1998-05-22 2005-11-29 Solexa, Inc. System and apparatus for sequential processing of analytes
US5919626A (en) 1997-06-06 1999-07-06 Orchid Bio Computer, Inc. Attachment of unmodified nucleic acids to silanized solid phase surfaces
WO1999005295A1 (en) 1997-07-25 1999-02-04 Thomas Jefferson University Composition and method for targeted integration into cells
WO1999009217A1 (en) 1997-08-15 1999-02-25 Hyseq, Inc. Methods and compositions for detection or quantification of nucleic acid species
GB9718455D0 (en) 1997-09-02 1997-11-05 Mcgregor Duncan P Chimeric binding peptide library screening method
JP2001519538A (en) 1997-10-10 2001-10-23 プレジデント・アンド・フェローズ・オブ・ハーバード・カレッジ Replica amplification of nucleic acid arrays
US6054274A (en) 1997-11-12 2000-04-25 Hewlett-Packard Company Method of amplifying the signal of target nucleic acid sequence analyte
US6242246B1 (en) 1997-12-15 2001-06-05 Somalogic, Inc. Nucleic acid ligand diagnostic Biochip
AU2003200718B2 (en) 1997-12-15 2006-10-19 Somalogic, Inc. Nucleic acid ligand diagnostic biochip
US6844158B1 (en) 1997-12-22 2005-01-18 Hitachi Chemical Co., Ltd. Direct RT-PCR on oligonucleotide-immobilized PCR microplates
EP1600512B1 (en) 1997-12-22 2008-05-28 Hitachi Chemical Co., Ltd. mRNA purification on microplates
US7427678B2 (en) 1998-01-08 2008-09-23 Sigma-Aldrich Co. Method for immobilizing oligonucleotides employing the cycloaddition bioconjugation method
DE69942272D1 (en) 1998-02-23 2010-06-02 Wisconsin Alumni Res Found DERANORDNUNGEN
WO1999044063A2 (en) 1998-02-25 1999-09-02 The United States Of America, Represented By The Secretary, Department Of Health And Human Services Tumor tissue microarrays for rapid molecular profiling
US6699710B1 (en) 1998-02-25 2004-03-02 The United States Of America As Represented By The Department Of Health And Human Services Tumor tissue microarrays for rapid molecular profiling
CA2318789C (en) 1998-02-25 2011-05-10 The United States Of America As Represented By The Secretary Department Of Health And Human Services Cellular arrays for rapid molecular profiling
WO1999053102A1 (en) 1998-04-16 1999-10-21 Packard Bioscience Company Analysis of polynucleotide sequence
US6906245B1 (en) 1998-04-30 2005-06-14 Sumitomo Chemical Company, Limited Method for producing transgenic plants resistant to weed control compounds which disrupt the porphyrin pathways of plants
US6358475B1 (en) 1998-05-27 2002-03-19 Becton, Dickinson And Company Device for preparing thin liquid for microscopic analysis
US6295153B1 (en) 1998-06-04 2001-09-25 Board Of Regents, The University Of Texas System Digital optical chemistry micromirror imager
AU754952B2 (en) 1998-06-24 2002-11-28 Illumina, Inc. Decoding of array sensors with microspheres
WO2000000637A2 (en) 1998-06-26 2000-01-06 Visible Genetics Inc. Method for sequencing nucleic acids with reduced errors
US20030022207A1 (en) 1998-10-16 2003-01-30 Solexa, Ltd. Arrayed polynucleotides and their use in genome analysis
US20040106110A1 (en) 1998-07-30 2004-06-03 Solexa, Ltd. Preparation of polynucleotide arrays
US6787308B2 (en) 1998-07-30 2004-09-07 Solexa Ltd. Arrayed biomolecules and their use in sequencing
CA2343359C (en) 1998-09-18 2008-12-23 Micromet Ag Dna amplification of a single cell
US6159736A (en) 1998-09-23 2000-12-12 Wisconsin Alumni Research Foundation Method for making insertional mutations using a Tn5 synaptic complex
AR021833A1 (en) 1998-09-30 2002-08-07 Applied Research Systems METHODS OF AMPLIFICATION AND SEQUENCING OF NUCLEIC ACID
US6573043B1 (en) 1998-10-07 2003-06-03 Genentech, Inc. Tissue analysis and kits therefor
US6337472B1 (en) 1998-10-19 2002-01-08 The University Of Texas System Board Of Regents Light imaging microscope having spatially resolved images
JP5213202B2 (en) 1998-10-28 2013-06-19 バイシス・インコーポレーテツド Detection and use of cell arrays and markers of genetic disorders
US6391937B1 (en) 1998-11-25 2002-05-21 Motorola, Inc. Polyacrylamide hydrogels and hydrogel arrays made from polyacrylamide reactive prepolymers
WO2000032823A1 (en) 1998-12-02 2000-06-08 Phylos, Inc. Dna-protein fusions and uses thereof
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
CA2355816C (en) 1998-12-14 2007-10-30 Li-Cor, Inc. A system and methods for nucleic acid sequencing of single molecules by polymerase synthesis
US6830884B1 (en) 1998-12-15 2004-12-14 Molecular Staging Inc. Method of amplification
EP1144684B1 (en) 1999-01-06 2009-08-19 Callida Genomics, Inc. Enhanced sequencing by hybridization using pools of probes
GB9901475D0 (en) 1999-01-22 1999-03-17 Pyrosequencing Ab A method of DNA sequencing
US6565727B1 (en) 1999-01-25 2003-05-20 Nanolytics, Inc. Actuators for microfluidics without moving parts
EP1024201B1 (en) 1999-01-27 2003-11-26 Commissariat A L'energie Atomique Microassay for serial analysis of gene expression and applications thereof
US20020150909A1 (en) 1999-02-09 2002-10-17 Stuelpnagel John R. Automated information processing in randomly ordered arrays
US6294063B1 (en) 1999-02-12 2001-09-25 Board Of Regents, The University Of Texas System Method and apparatus for programmable fluidic processing
US6153389A (en) 1999-02-22 2000-11-28 Haarer; Brian K. DNA additives as a mechanism for unambiguously marking biological samples
US20060275782A1 (en) 1999-04-20 2006-12-07 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
US20050191698A1 (en) 1999-04-20 2005-09-01 Illumina, Inc. Nucleic acid sequencing using microsphere arrays
CA2370976C (en) 1999-04-20 2009-10-20 Illumina, Inc. Detection of nucleic acid reactions on bead arrays
US6673620B1 (en) 1999-04-20 2004-01-06 Cytologix Corporation Fluid exchange in a chamber on a microscope slide
US6355431B1 (en) 1999-04-20 2002-03-12 Illumina, Inc. Detection of nucleic acid amplification reactions using bead arrays
EP1171636A2 (en) 1999-04-22 2002-01-16 Albert Einstein College Of Medicine Of Yeshiva University Assay of gene expression patterns by multi-fluor fish
US7276336B1 (en) 1999-07-22 2007-10-02 Agilent Technologies, Inc. Methods of fabricating an addressable array of biopolymer probes
US20010055764A1 (en) 1999-05-07 2001-12-27 Empedocles Stephen A. Microarray methods utilizing semiconductor nanocrystals
US6544732B1 (en) 1999-05-20 2003-04-08 Illumina, Inc. Encoding and decoding of array sensors utilizing nanocrystals
US6620584B1 (en) 1999-05-20 2003-09-16 Illumina Combinatorial decoding of random nucleic acid arrays
US6136592A (en) 1999-06-25 2000-10-24 Leighton; Stephen B. Multiple micro-arrays
US7501245B2 (en) 1999-06-28 2009-03-10 Helicos Biosciences Corp. Methods and apparatuses for analyzing polynucleotide sequences
US6818395B1 (en) 1999-06-28 2004-11-16 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences
US6465183B2 (en) 1999-07-01 2002-10-15 Agilent Technologies, Inc. Multidentate arrays
CA2379132C (en) 1999-07-14 2011-05-31 Packard Bioscience Company Method for multiplexed analysis of a plurality of target nucleic acid sequences in a sample
WO2001007915A2 (en) 1999-07-26 2001-02-01 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health & Human Services, The National Institutes Of Health Layered device with capture regions for cellular analysis
WO2001009363A1 (en) 1999-08-02 2001-02-08 Wisconsin Alumni Research Foundation Mutant tn5 transposase enzymes and method for their use
ATE318932T1 (en) 1999-08-13 2006-03-15 Univ Yale BINARY CODED SEQUENCE MARKERS
WO2001012862A2 (en) 1999-08-18 2001-02-22 Illumina, Inc. Compositions and methods for preparing oligonucleotide solutions
AU2246601A (en) 1999-08-30 2001-04-10 Illumina, Inc. Methods for improving signal detection from an array
KR100527265B1 (en) 1999-09-13 2005-11-09 뉴젠 테크놀로지스 인코포레이티드 Methods and compositions for linear isothermal amplification of polynucleotide sequences
US7244559B2 (en) 1999-09-16 2007-07-17 454 Life Sciences Corporation Method of sequencing a nucleic acid
US6274320B1 (en) 1999-09-16 2001-08-14 Curagen Corporation Method of sequencing a nucleic acid
US6544790B1 (en) 1999-09-17 2003-04-08 Whitehead Institute For Biomedical Research Reverse transfection method
US6291180B1 (en) 1999-09-29 2001-09-18 American Registry Of Pathology Ultrasound-mediated high-speed biological reaction and tissue processing
US6677160B1 (en) 1999-09-29 2004-01-13 Pharmacia & Upjohn Company Methods for creating a compound library and identifying lead chemical templates and ligands for target molecules
WO2001023610A2 (en) 1999-09-29 2001-04-05 Solexa Ltd. Polynucleotide sequencing
ATE432993T1 (en) 1999-10-04 2009-06-15 Univ New Jersey Med TAR RNA BINDING PEPTIDES
CA2386791A1 (en) 1999-10-08 2001-04-19 Protogene Laboratories, Inc. Method and apparatus for performing large numbers of reactions using array assembly
KR20020071853A (en) 1999-10-13 2002-09-13 시그나츄어 바이오사이언스 인코포레이티드 System and method for detecting and identifying molecular events in a test sample
US7585632B2 (en) 1999-10-29 2009-09-08 Hologic, Inc. Compositions and methods for the detection of a nucleic acid using a cleavage reaction
US6569674B1 (en) 1999-12-15 2003-05-27 Amersham Biosciences Ab Method and apparatus for performing biological reactions on a substrate surface
CA2394358A1 (en) 1999-12-13 2001-06-14 The Government Of The United States Of America, As Represented By The Se Cretary, Department Of Health & Human Services, The National Institutes High-throughput tissue microarray technology and applications
US6248535B1 (en) 1999-12-20 2001-06-19 University Of Southern California Method for isolation of RNA from formalin-fixed paraffin-embedded tissue specimens
US6485926B2 (en) 1999-12-22 2002-11-26 Fuji Photo Film Co., Ltd. Method for measuring protease activity
US6713298B2 (en) 2000-01-31 2004-03-30 Board Of Regents, The University Of Texas System Method and apparatus for the delivery of samples to a chemical sensor array
GB0002389D0 (en) 2000-02-02 2000-03-22 Solexa Ltd Molecular arrays
US7582420B2 (en) 2001-07-12 2009-09-01 Illumina, Inc. Multiplex nucleic acid reactions
US7611869B2 (en) 2000-02-07 2009-11-03 Illumina, Inc. Multiplexed methylation detection methods
US7361488B2 (en) 2000-02-07 2008-04-22 Illumina, Inc. Nucleic acid detection methods using universal priming
US8076063B2 (en) 2000-02-07 2011-12-13 Illumina, Inc. Multiplexed methylation detection methods
DE60127939T2 (en) 2000-02-07 2008-01-24 Illumina, Inc., San Diego Nucleic acid detection method with universal priming
US7955794B2 (en) 2000-09-21 2011-06-07 Illumina, Inc. Multiplex nucleic acid reactions
ATE411397T1 (en) 2000-02-07 2008-10-15 Illumina Inc NUCLEIC ACID DETECTION METHOD WITH UNIVERSAL PRIMING
US20010026919A1 (en) 2000-02-08 2001-10-04 Alex Chenchik Nucleic acid assays employing universal arrays
US6770441B2 (en) 2000-02-10 2004-08-03 Illumina, Inc. Array compositions and methods of making same
AU3839101A (en) 2000-02-15 2001-08-27 Lynx Therapeutics, Inc. Data analysis and display system for ligation-based dna sequencing
US7306904B2 (en) 2000-02-18 2007-12-11 Olink Ab Methods and kits for proximity probing
JP2001284267A (en) 2000-04-03 2001-10-12 Canon Inc Exhaust gas processing method, and plasma processing method and apparatus
WO2001077684A2 (en) 2000-04-10 2001-10-18 The Scripps Research Institute Proteomic analysis using activity-based probe libraries
US6368801B1 (en) 2000-04-12 2002-04-09 Molecular Staging, Inc. Detection and amplification of RNA using target-mediated ligation of DNA by RNA ligase
US7001792B2 (en) 2000-04-24 2006-02-21 Eagle Research & Development, Llc Ultra-fast nucleic acid sequencing device and a method for making and using the same
US6291187B1 (en) 2000-05-12 2001-09-18 Molecular Staging, Inc. Poly-primed amplification of nucleic acid sequences
AU2001274869A1 (en) 2000-05-20 2001-12-03 The Regents Of The University Of Michigan Method of producing a dna library using positional amplification
US6511809B2 (en) 2000-06-13 2003-01-28 E. I. Du Pont De Nemours And Company Method for the detection of an analyte by means of a nucleic acid reporter
US7439016B1 (en) 2000-06-15 2008-10-21 Digene Corporation Detection of nucleic acids by type-specific hybrid capture method
JP4744778B2 (en) 2000-06-21 2011-08-10 バイオアレイ ソルーションズ リミテッド Method for analyzing multiple analyte molecules using a specific random particle array
US8278034B2 (en) 2000-06-22 2012-10-02 Nuclea Biotechnologies, Inc. Methods of making frozen tissue microarrays
US6323009B1 (en) 2000-06-28 2001-11-27 Molecular Staging, Inc. Multiply-primed amplification of nucleic acid sequences
DE60131194T2 (en) 2000-07-07 2008-08-07 Visigen Biotechnologies, Inc., Bellaire SEQUENCE PROVISION IN REAL TIME
GB0018120D0 (en) 2000-07-24 2000-09-13 Fermentas Ab Nuclease
WO2002007503A1 (en) 2000-07-25 2002-01-31 The Regents Of The University Of California Electrowetting-driven micropumping
US7613571B2 (en) 2000-07-28 2009-11-03 Doyle Michael D Method and system for the multidimensional morphological reconstruction of genome expression activity
ATE330224T1 (en) 2000-08-15 2006-07-15 Discerna Ltd FUNCTIONAL PROTEIN ARRAY
SI1918298T1 (en) 2000-08-21 2011-01-31 Apitope Technology Bristol Ltd Peptide
US6713257B2 (en) 2000-08-25 2004-03-30 Rosetta Inpharmatics Llc Gene discovery using microarrays
US6773566B2 (en) 2000-08-31 2004-08-10 Nanolytics, Inc. Electrostatic actuators for microfluidics and methods for using same
US6942970B2 (en) 2000-09-14 2005-09-13 Zymed Laboratories, Inc. Identifying subjects suitable for topoisomerase II inhibitor treatment
US20020168639A1 (en) 2000-09-22 2002-11-14 Muraca Patrick J. Profile array substrates
AU2001293163A1 (en) 2000-09-27 2002-04-08 Lynx Therapeutics, Inc. Method for determining relative abundance of nucleic acid sequences
AT411066B (en) 2000-10-24 2003-09-25 Steiner Georg E METHOD AND ARRANGEMENT FOR THE INVESTIGATION OF CELLS
EP1348034B1 (en) 2000-11-15 2016-07-20 Minerva Biotechnologies Corporation Oligonucleotide identifiers
EP2381116A1 (en) 2000-11-16 2011-10-26 California Institute of Technology Apparatus and methods for conducting assays and high throughput screening
WO2002044425A2 (en) 2000-12-01 2002-06-06 Visigen Biotechnologies, Inc. Enzymatic nucleic acid synthesis: compositions and methods for altering monomer incorporation fidelity
AR031640A1 (en) 2000-12-08 2003-09-24 Applied Research Systems ISOTHERMAL AMPLIFICATION OF NUCLEIC ACIDS IN A SOLID SUPPORT
US20030215936A1 (en) 2000-12-13 2003-11-20 Olli Kallioniemi High-throughput tissue microarray technology and applications
US20030017451A1 (en) 2000-12-21 2003-01-23 Hui Wang Methods for detecting transcripts
US7135296B2 (en) * 2000-12-28 2006-11-14 Mds Inc. Elemental analysis of tagged biologically active materials
JP4061043B2 (en) 2000-12-28 2008-03-12 株式会社ポストゲノム研究所 Method for producing peptide etc. by in vitro transcription / translation system
ATE538380T1 (en) 2001-01-23 2012-01-15 Harvard College NUCLEIC ACID PROGRAMMABLE PROTEIN ARRAYS
EP1356117A2 (en) 2001-01-25 2003-10-29 TM Bioscience Corporation Polynucleotides for use as tags and tag complements, manufacture and use thereof
US20030087232A1 (en) 2001-01-25 2003-05-08 Fred Christians Methods for screening polypeptides
KR20020063359A (en) 2001-01-27 2002-08-03 일렉트론 바이오 (주) nucleic hybridization assay method and device using a cleavage technique responsive to the specific sequences of the complementary double strand of nucleic acids or oligonucleotides
EP1379549A2 (en) 2001-02-07 2004-01-14 Institut Pasteur Sequence of the photorhabdus luminescens strain tt01 genome and uses
US6573051B2 (en) 2001-03-09 2003-06-03 Molecular Staging, Inc. Open circle probes with intramolecular stem structures
WO2002072892A1 (en) 2001-03-12 2002-09-19 California Institute Of Technology Methods and apparatus for analyzing polynucleotide sequences by asynchronous base extension
AU785425B2 (en) 2001-03-30 2007-05-17 Genetic Technologies Limited Methods of genomic analysis
ES2400448T3 (en) 2001-04-30 2013-04-10 Ventana Medical Systems, Inc. Reagents and methods for automated in situ hybridization or microarray
WO2003002979A2 (en) 2001-06-28 2003-01-09 Illumina, Inc. Multiplex decoding of array sensors with microspheres
US7473767B2 (en) 2001-07-03 2009-01-06 The Institute For Systems Biology Methods for detection and quantification of analytes in complex mixtures
WO2003008968A2 (en) 2001-07-19 2003-01-30 Signet Laboratories, Inc. Human tissue specific drug screening procedure
US20040091857A1 (en) 2001-07-20 2004-05-13 Nallur Girish N. Gene expression profiling
GB0118031D0 (en) 2001-07-24 2001-09-19 Oxford Glycosciences Uk Ltd Self assembled protein nucleic acid complexes and self assembled protein arrays
US7297778B2 (en) 2001-07-25 2007-11-20 Affymetrix, Inc. Complexity management of genomic DNA
DE60210014T2 (en) 2001-07-31 2006-09-21 Pfizer Products Inc., Groton Cell-based phosphodiesterase 10A assay and sequences
US6696271B2 (en) 2001-08-23 2004-02-24 The Regents Of The University Of California Frozen tissue microarray technology for analysis of RNA, DNA, and proteins
WO2003022028A2 (en) 2001-09-10 2003-03-20 Meso Scale Technologies, Llc Methods, reagents, kits and apparatus for protein function analysis
US20060188875A1 (en) 2001-09-18 2006-08-24 Perlegen Sciences, Inc. Human genomic polymorphisms
US20040019005A1 (en) 2001-10-01 2004-01-29 Jeffrey Van Ness Methods for parallel measurement of genetic variations
US7195913B2 (en) 2001-10-05 2007-03-27 Surmodics, Inc. Randomly ordered arrays and methods of making and using
US20030148335A1 (en) 2001-10-10 2003-08-07 Li Shen Detecting targets by unique identifier nucleotide tags
US6942972B2 (en) 2001-10-24 2005-09-13 Beckman Coulter, Inc. Efficient synthesis of protein-oligonucleotide conjugates
US20030175947A1 (en) 2001-11-05 2003-09-18 Liu Robin Hui Enhanced mixing in microfluidic devices
AU2002365271B2 (en) 2001-11-06 2008-12-11 Agilix Corporation Sensitive coded detection systems
EP1451365A4 (en) 2001-11-13 2006-09-13 Rubicon Genomics Inc Dna amplification and sequencing using dna molecules generated by random fragmentation
GB0127564D0 (en) 2001-11-16 2002-01-09 Medical Res Council Emulsion compositions
EP1450956A2 (en) 2001-11-26 2004-09-01 Keck Graduate Institute Method, apparatus and article for microfluidic control via electrowetting, for chemical, biochemical and biological assays and the like
US7057026B2 (en) 2001-12-04 2006-06-06 Solexa Limited Labelled nucleotides
US7098041B2 (en) 2001-12-11 2006-08-29 Kimberly-Clark Worldwide, Inc. Methods to view and analyze the results from diffraction-based diagnostics
US20030153850A1 (en) 2002-01-16 2003-08-14 Davis Brian J. Method and apparatus for image-guided therapy
US7731909B1 (en) 2002-01-22 2010-06-08 Grace Bio-Labs, Inc. Reaction surface array diagnostic apparatus
WO2003069333A1 (en) 2002-02-14 2003-08-21 Illumina, Inc. Automated information processing in randomly ordered arrays
US20040002090A1 (en) 2002-03-05 2004-01-01 Pascal Mayer Methods for detecting genome-wide sequence variations associated with a phenotype
US20030170637A1 (en) 2002-03-06 2003-09-11 Pirrung Michael C. Method of analyzing mRNA splice variants
US7223371B2 (en) 2002-03-14 2007-05-29 Micronics, Inc. Microfluidic channel network device
AU2003228531A1 (en) 2002-04-15 2003-11-03 The Regents Of The University Of California Screening and therapeutic methods for treating circadian rhythm disorders
US20030235851A1 (en) 2002-04-19 2003-12-25 Roberts Richard W. Methods of using sense and/or nonsense suppression to make nucleic acid-peptide display libraries containing peptides with unnatural amino acid residues
PT2151250E (en) 2002-05-06 2013-12-09 Endocyte Inc Vitamin-targeted imaging agents
EP1540017A4 (en) 2002-05-09 2006-05-24 Us Genomics Inc Methods for analyzing a nucleic acid
ES2346646T5 (en) 2002-05-30 2018-02-15 The Scripps Research Institute Copper catalyzed ligation of azides and acetylenes
WO2003102233A1 (en) 2002-06-03 2003-12-11 Pamgene B.V. Novel high density arrays and methods for analyte analysis
US7108976B2 (en) 2002-06-17 2006-09-19 Affymetrix, Inc. Complexity management of genomic DNA by locus specific amplification
FR2841063B1 (en) 2002-06-18 2004-09-17 Commissariat Energie Atomique DEVICE FOR DISPLACING SMALL VOLUMES OF LIQUID ALONG A MICRO-CATENARY BY ELECTROSTATIC FORCES
JP2005530151A (en) 2002-06-18 2005-10-06 インヴィトロジェン コーポレーション Method and apparatus for low resistance electrophoresis of precast hydratable separation media
US20050019776A1 (en) 2002-06-28 2005-01-27 Callow Matthew James Universal selective genome amplification and universal genotyping system
US7205128B2 (en) 2002-08-16 2007-04-17 Agilent Technologies, Inc. Method for synthesis of the second strand of cDNA
US20050118616A1 (en) 2002-08-16 2005-06-02 Kawashima Tadashi R. Amplification of target nucleotide sequence without polymerase chain reaction
EP1530578B1 (en) 2002-08-23 2013-03-13 Illumina Cambridge Limited Modified nucleotides for polynucleotide sequencing
US20040038385A1 (en) 2002-08-26 2004-02-26 Langlois Richard G. System for autonomous monitoring of bioagents
US20070166725A1 (en) 2006-01-18 2007-07-19 The Regents Of The University Of California Multiplexed diagnostic platform for point-of care pathogen detection
CA2498362C (en) 2002-09-11 2012-11-06 Temple University - Of The Commonwealth System Of Higher Education Automated system for high-throughput electrophoretic separations
US7595883B1 (en) 2002-09-16 2009-09-29 The Board Of Trustees Of The Leland Stanford Junior University Biological analysis arrangement and approach therefor
JP4632788B2 (en) 2002-09-20 2011-02-16 ニュー・イングランド・バイオラブズ・インコーポレイティッド Helicase-dependent amplification of nucleic acids
US7662594B2 (en) 2002-09-20 2010-02-16 New England Biolabs, Inc. Helicase-dependent amplification of RNA
US6911132B2 (en) 2002-09-24 2005-06-28 Duke University Apparatus for manipulating droplets by electrowetting-based techniques
WO2004028955A2 (en) 2002-09-25 2004-04-08 California Institute Of Technology Microfluidic large scale integration
US20040259105A1 (en) 2002-10-03 2004-12-23 Jian-Bing Fan Multiplex nucleic acid analysis using archived or fixed samples
AU2003291632A1 (en) 2002-10-03 2004-04-23 Epimmune Inc Hla binding peptides and their uses
US20040067492A1 (en) 2002-10-04 2004-04-08 Allan Peng Reverse transcription on microarrays
WO2004041994A1 (en) 2002-10-31 2004-05-21 University Of Massachusetts Rapid cell block embedding method and apparatus
US7122384B2 (en) 2002-11-06 2006-10-17 E. I. Du Pont De Nemours And Company Resonant light scattering microparticle methods
US7132233B2 (en) 2002-12-12 2006-11-07 Novartis Vaccines And Diagnostics, Inc. Identification of oligonucleotides for the capture, detection and quantitation of West Nile virus
AU2003301061A1 (en) 2002-12-18 2004-07-22 West Virginia University Research Corporation Apparatus and method for edman degradation using a microfluidic system
KR20040062847A (en) 2003-01-03 2004-07-09 삼성전자주식회사 Method for replicating nucleic acid array
US7547380B2 (en) 2003-01-13 2009-06-16 North Carolina State University Droplet transportation devices and methods having a fluid surface
ES2342665T3 (en) 2003-01-29 2010-07-12 454 Corporation SEQUENCING FROM TWO EXTREME.
GB0302058D0 (en) 2003-01-29 2003-02-26 Univ Cranfield Replication of nucleic acid arrays
EP2365095A1 (en) 2003-02-26 2011-09-14 Callida Genomics, Inc. Random array DNA analysis by hybridization
US7718403B2 (en) 2003-03-07 2010-05-18 Rubicon Genomics, Inc. Amplification and analysis of whole genome and whole transcriptome libraries generated by a DNA polymerization process
EP1601450B1 (en) 2003-03-10 2013-06-05 Expression Pathology, Inc. Liquid tissue preparation from histopatologically processed biological samples, tissues and cells
FR2852317B1 (en) 2003-03-13 2006-08-04 PROBE BIOPUCES AND METHODS OF USE
US7083980B2 (en) 2003-04-17 2006-08-01 Wisconsin Alumni Research Foundation Tn5 transposase mutants and the use thereof
CN1300333C (en) 2003-04-17 2007-02-14 中国人民解放军军事医学科学院放射与辐射医学研究所 Preparation of gene chip for digagnosingantrax baiuus and its application
US7267994B2 (en) * 2003-04-28 2007-09-11 Regents Of The University Of California Element-coded affinity tags
US20040219588A1 (en) 2003-04-30 2004-11-04 Masaru Furuta Method for dispensing reagents onto biological samples and method for analyzing biological samples
US7666612B2 (en) 2003-05-23 2010-02-23 Epfl-Ecole Polytechnique Federale De Lausanne Methods for protein labeling based on acyl carrier protein
US20050026188A1 (en) 2003-05-30 2005-02-03 Van Kessel Andrew G. Methods of identifying, characterizing and comparing organism communities
KR100706464B1 (en) 2003-05-30 2007-04-10 애플라 코포레이션 Apparatus and method for hybridization and spr detection
US7255994B2 (en) 2003-06-10 2007-08-14 Applera Corporation Ligation assay
US20060216775A1 (en) 2003-06-17 2006-09-28 The Regents Of The University Of Califoenia Compositions and methods for analysis and manipulation of enzymes in biosynthetic proteomes
US20050053980A1 (en) 2003-06-20 2005-03-10 Illumina, Inc. Methods and compositions for whole genome amplification and genotyping
US20070128656A1 (en) 2003-06-26 2007-06-07 University Of South Florida Direct Fluorescent Label Incorporation Via 1st Strand cDNA Synthesis
US7601492B2 (en) 2003-07-03 2009-10-13 The Regents Of The University Of California Genome mapping of functional DNA elements and cellular proteins
JP5183063B2 (en) 2003-07-05 2013-04-17 ザ ジョンズ ホプキンス ユニバーシティ Methods and compositions for detection and enumeration of genetic variations
US20050079520A1 (en) 2003-07-21 2005-04-14 Jie Wu Multiplexed analyte detection
US20050031176A1 (en) 2003-08-08 2005-02-10 Hertel Sarah R. Method and apparatus of multi-modality image fusion
AU2004265635A1 (en) 2003-08-08 2005-02-24 Thomas Jefferson University Method for rapid identification of alternative splicing
US20050037362A1 (en) 2003-08-11 2005-02-17 Eppendorf Array Technologies, S.A. Detection and quantification of siRNA on microarrays
WO2005021794A2 (en) 2003-09-02 2005-03-10 Keygene N.V. Ola-based methods for the detection of target nucleic acid sequences
US20050095627A1 (en) 2003-09-03 2005-05-05 The Salk Institute For Biological Studies Multiple antigen detection assays and reagents
US7824856B2 (en) 2003-09-10 2010-11-02 Althea Technologies, Inc. Expression profiling using microarrays
GB0321306D0 (en) 2003-09-11 2003-10-15 Solexa Ltd Modified polymerases for improved incorporation of nucleotide analogues
US20050266417A1 (en) 2003-09-12 2005-12-01 Francis Barany Methods for identifying target nucleic acid molecules
DK1670939T3 (en) 2003-09-18 2010-03-01 Nuevolution As Method for obtaining structural information on a coded molecule and method for selecting compounds
US7541166B2 (en) 2003-09-19 2009-06-02 Microfluidic Systems, Inc. Sonication to selectively lyse different cell types
US20050064435A1 (en) 2003-09-24 2005-03-24 Xing Su Programmable molecular barcodes
US7328979B2 (en) 2003-11-17 2008-02-12 Koninklijke Philips Electronics N.V. System for manipulation of a body of fluid
WO2005051984A2 (en) 2003-11-21 2005-06-09 Arena Pharmaceuticals, Inc. Methods for producing olfactory gpcrs
CA2545006C (en) 2003-12-12 2013-09-17 Saint Louis University Biosensors for detecting macromolecules and other analytes
US7259258B2 (en) 2003-12-17 2007-08-21 Illumina, Inc. Methods of attaching biological compounds to solid supports using triazine
US20050136414A1 (en) 2003-12-23 2005-06-23 Kevin Gunderson Methods and compositions for making locus-specific arrays
US20050147976A1 (en) 2003-12-29 2005-07-07 Xing Su Methods for determining nucleotide sequence information
US20070014810A1 (en) 2003-12-31 2007-01-18 Denise Baker Inducing cellular immune responses to human papillomavirus using peptide and nucleic acid compositions
EP3175914A1 (en) 2004-01-07 2017-06-07 Illumina Cambridge Limited Improvements in or relating to molecular arrays
US7569392B2 (en) 2004-01-08 2009-08-04 Vanderbilt University Multiplex spatial profiling of gene expression
WO2005067648A2 (en) 2004-01-08 2005-07-28 Vanderbilt University Multiplex spatial profiling of gene expression
WO2005071110A2 (en) 2004-01-23 2005-08-04 Lingvitae As Improving polynucleotide ligation reactions
FR2866493B1 (en) 2004-02-16 2010-08-20 Commissariat Energie Atomique DEVICE FOR CONTROLLING THE DISPLACEMENT OF A DROP BETWEEN TWO OR MORE SOLID SUBSTRATES
US7378242B2 (en) 2004-03-18 2008-05-27 Atom Sciences, Inc. DNA sequence detection by limited primer extension
FR2868638B1 (en) 2004-03-30 2006-05-19 Sagem METHOD OF EXCHANGING INFORMATION BETWEEN TWO NETWORKS OPERATING UNDER DIFFERENT ROUTING PROTOCOLS
KR100624420B1 (en) 2004-04-10 2006-09-19 삼성전자주식회사 A microarray having microarray identification information stored in the form of a spot, method of producing the microarray and method of using the microarray
US20050260653A1 (en) 2004-04-14 2005-11-24 Joshua Labaer Nucleic-acid programmable protein arrays
JP4592060B2 (en) 2004-04-26 2010-12-01 キヤノン株式会社 PCR amplification reaction apparatus and PCR amplification reaction method using the apparatus
RU2270254C2 (en) 2004-04-30 2006-02-20 Институт Молекулярной Биологии Им. В.А. Энгельгардта Российской Академии Наук Identification of transgenic dna sequences in plant material and products made of the same, oligonucleotide kit and bioarray therefor
DE102004022263A1 (en) 2004-05-06 2005-12-15 Clondiag Chip Technologies Gmbh Apparatus and method for detecting molecular interactions
EP1756307A1 (en) 2004-05-20 2007-02-28 Trillion Genomics Limited Use of mass labelled probes to detect target nucleic acids using mass spectrometry
US7906276B2 (en) 2004-06-30 2011-03-15 Kimberly-Clark Worldwide, Inc. Enzymatic detection techniques
FR2872715B1 (en) 2004-07-08 2006-11-17 Commissariat Energie Atomique MICROREACTOR DROP
FR2872809B1 (en) 2004-07-09 2006-09-15 Commissariat Energie Atomique METHOD OF ADDRESSING ELECTRODES
US7608434B2 (en) 2004-08-04 2009-10-27 Wisconsin Alumni Research Foundation Mutated Tn5 transposase proteins and the use thereof
WO2006073504A2 (en) 2004-08-04 2006-07-13 President And Fellows Of Harvard College Wobble sequencing
JP2006058031A (en) 2004-08-17 2006-03-02 Hitachi High-Technologies Corp Chemical analyzer
US7776547B2 (en) 2004-08-26 2010-08-17 Intel Corporation Cellular analysis using Raman surface scanning
US20060228758A1 (en) 2004-09-13 2006-10-12 Xencor, Inc. Analysis of MHC-peptide binding interactions
CN100396790C (en) 2004-09-17 2008-06-25 北京大学 Solution identification and surface addressing protein chip and its preparing and detecting method
GB2423819B (en) 2004-09-17 2008-02-06 Pacific Biosciences California Apparatus and method for analysis of molecules
US7524672B2 (en) 2004-09-22 2009-04-28 Sandia Corporation Microfluidic microarray systems and methods thereof
US7527970B2 (en) 2004-10-15 2009-05-05 The United States Of America As Represented By The Department Of Health And Human Services Method of identifying active chromatin domains
EP1816192B1 (en) 2004-10-15 2012-12-12 National Institute of Advanced Industrial Science and Technology LINKER FOR CONSTRUCTING mRNA-PUROMYCIN-PROTEIN CONJUGATE
EP2322616A1 (en) 2004-11-12 2011-05-18 Asuragen, Inc. Methods and compositions involving miRNA and miRNA inhibitor molecules
US7183119B2 (en) 2004-11-15 2007-02-27 Eastman Kodak Company Method for sensitive detection of multiple biological analytes
US7745143B2 (en) 2004-11-19 2010-06-29 Plexera, Llc Plasmon resonance biosensor and method
DK1828381T3 (en) 2004-11-22 2009-02-23 Peter Birk Rasmussen Template-directed split-and-mix synthesis of small molecule libraries
ITPD20040301A1 (en) 2004-11-26 2005-02-26 Dimensional Srl P METHOD AND APPARATUS FOR THE SIMULTANEOUS SEPARATION OF BIOLOGICAL MOLECULES BY BIDIMENSIONAL ELECTROPHORESIS
WO2006064199A1 (en) 2004-12-13 2006-06-22 Solexa Limited Improved method of nucleotide detection
GB0427236D0 (en) 2004-12-13 2005-01-12 Solexa Ltd Improved method of nucleotide detection
US20060292586A1 (en) 2004-12-17 2006-12-28 Schroth Gary P ID-tag complexes, arrays, and methods of use thereof
WO2006074351A2 (en) 2005-01-05 2006-07-13 Agencourt Personal Genomics Reversible nucleotide terminators and uses thereof
KR100682920B1 (en) 2005-01-20 2007-02-15 삼성전자주식회사 Microfluidic chip for multiple bioassay and its method for production
WO2006081222A2 (en) 2005-01-25 2006-08-03 Compass Genetics, Llc. Isothermal dna amplification
US7458661B2 (en) 2005-01-25 2008-12-02 The Regents Of The University Of California Method and apparatus for promoting the complete transfer of liquid drops from a nozzle
CN101916359B (en) 2005-01-27 2012-06-20 剑桥研究和仪器设备股份有限公司 Methods and apparatus for classifying different parts of a sample into respective classes
EP1859330B1 (en) 2005-01-28 2012-07-04 Duke University Apparatuses and methods for manipulating droplets on a printed circuit board
EP2230315A1 (en) 2005-02-01 2010-09-22 AB Advanced Genetic Analysis Corporation Nucleic acid sequencing by performing successive cycles of duplex extension
JP2008529035A (en) 2005-02-03 2008-07-31 パーキンエルマー エルエーエス,インク. Ultra-sensitive detection system using multidimensional signals
US7393665B2 (en) 2005-02-10 2008-07-01 Population Genetics Technologies Ltd Methods and compositions for tagging and identifying polynucleotides
US7407757B2 (en) 2005-02-10 2008-08-05 Population Genetics Technologies Genetic analysis by sequence-specific sorting
US20060199207A1 (en) 2005-02-24 2006-09-07 Matysiak Stefan M Self-assembly of molecules using combinatorial hybridization
US7727721B2 (en) 2005-03-08 2010-06-01 California Institute Of Technology Hybridization chain reaction amplification for in situ imaging
GB0504774D0 (en) 2005-03-08 2005-04-13 Lingvitae As Method
US7776567B2 (en) 2005-03-17 2010-08-17 Biotium, Inc. Dimeric and trimeric nucleic acid dyes, and associated systems and methods
US7601498B2 (en) 2005-03-17 2009-10-13 Biotium, Inc. Methods of using dyes in association with nucleic acid staining or detection and associated technology
US7303880B2 (en) 2005-03-18 2007-12-04 Wisconsin Alumni Research Foundation Microdissection-based methods for determining genomic features of single chromosomes
US7229769B2 (en) 2005-03-25 2007-06-12 Illumina, Inc. Compositions and methods for detecting protease activity
ES2313143T3 (en) 2005-04-06 2009-03-01 Maurice Stroun METHOD FOR THE CANCER DIAGNOSIS THROUGH CIRCULATING DNA AND RNA DETECTION.
GB0508983D0 (en) 2005-05-03 2005-06-08 Oxford Gene Tech Ip Ltd Cell analyser
JP4990886B2 (en) 2005-05-10 2012-08-01 ソレックサ リミテッド Improved polymerase
WO2006124644A2 (en) 2005-05-12 2006-11-23 Board Of Regents, The University Of Texas System Protein and antibody profiling using small molecule microarrays
ATE502120T1 (en) 2005-05-12 2011-04-15 Affymetrix Inc MULTIPLEX ASSAY FOR BRANCHED CHAIN DNA
GB0509833D0 (en) 2005-05-16 2005-06-22 Isis Innovation Cell analysis
CA2608751A1 (en) 2005-05-18 2006-11-23 Novartis Ag Methods for diagnosis and treatment of proliferative disorders mediated by cd40 signaling
US20060263789A1 (en) 2005-05-19 2006-11-23 Robert Kincaid Unique identifiers for indicating properties associated with entities to which they are attached, and methods for using
CN100526453C (en) 2005-05-20 2009-08-12 麦克奥迪实业集团有限公司 Cell collection method after laser microdissection
CA2609328A1 (en) 2005-05-26 2006-11-30 The Trustees Of Boston University Quantification of nucleic acids and proteins using oligonucleotide mass tags
US8486629B2 (en) 2005-06-01 2013-07-16 Bioarray Solutions, Ltd. Creation of functionalized microparticle libraries by oligonucleotide ligation or elongation
EP1910537A1 (en) 2005-06-06 2008-04-16 454 Life Sciences Corporation Paired end sequencing
EP1889047B1 (en) 2005-06-07 2011-08-10 Centre National De La Recherche Scientifique -Cnrs- Use of conjugates with linkers cleavable by photodissociation or fragmentation for mass spectrometry analysis of tissue sections
GB0511717D0 (en) 2005-06-09 2005-07-13 Babraham Inst Repeatable protein arrays
CA2611671C (en) 2005-06-15 2013-10-08 Callida Genomics, Inc. Single molecule arrays for genetic and chemical analysis
US7709198B2 (en) 2005-06-20 2010-05-04 Advanced Cell Diagnostics, Inc. Multiplex detection of nucleic acids
US20070087360A1 (en) 2005-06-20 2007-04-19 Boyd Victoria L Methods and compositions for detecting nucleotides
US7873193B2 (en) 2005-06-21 2011-01-18 Carl Zeiss Microimaging Gmbh Serial section analysis for computer-controlled microscopic imaging
DE602006018744D1 (en) 2005-06-23 2011-01-20 Keygene Nv STRATEGIES WITH HIGH THROUGHPUT TO IDENTIFY AND DETECT POLYMORPHISMS
US20070026430A1 (en) 2005-06-30 2007-02-01 Applera Corporation Proximity probing of target proteins comprising restriction and/or extension
US7883848B2 (en) 2005-07-08 2011-02-08 Olink Ab Regulation analysis by cis reactivity, RACR
JP4822753B2 (en) 2005-07-11 2011-11-24 一般社団法人オンチップ・セロミクス・コンソーシアム Cell component sorting chip, cell component analysis system, and cell component analysis method using them
GB0514936D0 (en) 2005-07-20 2005-08-24 Solexa Ltd Preparation of templates for nucleic acid sequencing
US20070020640A1 (en) 2005-07-21 2007-01-25 Mccloskey Megan L Molecular encoding of nucleic acid templates for PCR and other forms of sequence analysis
US20070023292A1 (en) 2005-07-26 2007-02-01 The Regents Of The University Of California Small object moving on printed circuit board
US7805081B2 (en) 2005-08-11 2010-09-28 Pacific Biosciences Of California, Inc. Methods and systems for monitoring multiple optical signals from a single source
WO2007027653A1 (en) 2005-09-01 2007-03-08 Promega Corporation Cell-based luminogenic and nonluminogenic proteasome assays
WO2007030373A2 (en) 2005-09-07 2007-03-15 St. Jude Children's Research Hospital Method for in situ hybridization analysis
JP2007074967A (en) 2005-09-13 2007-03-29 Canon Inc Identifier probe and method for amplifying nucleic acid by using the same
US7405281B2 (en) 2005-09-29 2008-07-29 Pacific Biosciences Of California, Inc. Fluorescent nucleotide analogs and uses therefor
CA2623539C (en) 2005-09-29 2015-12-15 Keygene N.V. High throughput screening of mutagenized populations
WO2007041689A2 (en) 2005-10-04 2007-04-12 President And Fellows Of Harvard College Methods of site-specific labeling of molecules and molecules produced thereby
GB0522310D0 (en) 2005-11-01 2005-12-07 Solexa Ltd Methods of preparing libraries of template polynucleotides
US20070116612A1 (en) 2005-11-02 2007-05-24 Biopath Automation, L.L.C. Prefix tissue cassette
EP1951905B1 (en) 2005-11-10 2012-04-11 Affymetrix, Inc. Detection of nucleic acids through amplification of surrogate nucleic acids
WO2007120208A2 (en) 2005-11-14 2007-10-25 President And Fellows Of Harvard College Nanogrid rolling circle dna sequencing
ATE527383T1 (en) 2005-11-22 2011-10-15 Stichting Dienst Landbouwkundi MULTIPLEX NUCLEIC ACID DETECTION
EP1957979A1 (en) 2005-11-25 2008-08-20 Koninklijke Philips Electronics N.V. Magnetic biosensor for determination of enzymic activity
WO2007100392A2 (en) 2005-11-30 2007-09-07 Biotium, Inc. Enzyme substrate comprising a functional dye and associated technology and methods
US20070161029A1 (en) 2005-12-05 2007-07-12 Panomics, Inc. High throughput profiling of methylation status of promoter regions of genes
US7803751B2 (en) 2005-12-09 2010-09-28 Illumina, Inc. Compositions and methods for detecting phosphomonoester
DE102005060738A1 (en) 2005-12-16 2007-06-21 Qiagen Gmbh Method for extraction of biomolecules from fixed tissues
US20070141718A1 (en) 2005-12-19 2007-06-21 Bui Huy A Reduction of scan time in imaging mass spectrometry
US20070178503A1 (en) 2005-12-19 2007-08-02 Feng Jiang In-situ genomic DNA chip for detection of cancer
DK3404114T3 (en) 2005-12-22 2021-06-28 Keygene Nv Method for detecting high throughput AFLP-based polymorphism
ES2394633T3 (en) 2005-12-22 2013-02-04 Keygene N.V. Improved strategies for developing transcript profiles using high performance sequencing technologies
AU2006330834B2 (en) 2005-12-23 2013-09-12 Nanostring Technologies, Inc. Compositions comprising oriented, immobilized macromolecules and methods for their preparation
EP1985092B1 (en) 2005-12-23 2011-07-06 Telefonaktiebolaget LM Ericsson (publ) Method and apparatus for solving data packet traffic congestion.
US20100015607A1 (en) 2005-12-23 2010-01-21 Nanostring Technologies, Inc. Nanoreporters and methods of manufacturing and use thereof
WO2007076726A1 (en) 2006-01-04 2007-07-12 Si Lok Methods for nucleic acid mapping and identification of fine-structural-variations in nucleic acids and utilities
US7544473B2 (en) 2006-01-23 2009-06-09 Population Genetics Technologies Ltd. Nucleic acid analysis using sequence tokens
WO2007087312A2 (en) 2006-01-23 2007-08-02 Population Genetics Technologies Ltd. Molecular counting
WO2007087339A2 (en) 2006-01-24 2007-08-02 Perkinelmer Las, Inc. Multiplexed analyte quantitation by two-dimensional planar electrochromatography
WO2007092538A2 (en) 2006-02-07 2007-08-16 President And Fellows Of Harvard College Methods for making nucleotide probes for sequencing and synthesis
JP5180845B2 (en) 2006-02-24 2013-04-10 カリダ・ジェノミックス・インコーポレイテッド High-throughput genomic sequencing on DNA arrays
SG10201405158QA (en) 2006-02-24 2014-10-30 Callida Genomics Inc High throughput genome sequencing on dna arrays
CN101395280A (en) 2006-03-01 2009-03-25 凯津公司 High throughput sequence-based detection of snps using ligation assays
EP2021503A1 (en) 2006-03-17 2009-02-11 Solexa Ltd. Isothermal methods for creating clonal single molecule arrays
JP2007248396A (en) 2006-03-17 2007-09-27 Toshiba Corp Device for detecting nucleic acid, and nucleic acid detector
GB0605584D0 (en) 2006-03-20 2006-04-26 Olink Ab Method for analyte detection using proximity probes
US20070231823A1 (en) 2006-03-23 2007-10-04 Mckernan Kevin J Directed enrichment of genomic DNA for high-throughput sequencing
US8975216B2 (en) 2006-03-30 2015-03-10 Pacific Biosciences Of California Articles having localized molecules disposed thereon and methods of producing same
CN101460953B (en) 2006-03-31 2012-05-30 索雷克萨公司 Systems and devices for sequence by synthesis analysis
DK3239304T3 (en) 2006-04-04 2020-10-26 Keygene Nv High-throughput detection of molecular markers based on AFLP and high-throughput sequencing
JP5054096B2 (en) 2006-04-18 2012-10-24 アドヴァンスト リキッド ロジック インコーポレイテッド Biochemistry based on droplets
US7439014B2 (en) 2006-04-18 2008-10-21 Advanced Liquid Logic, Inc. Droplet-based surface modification and washing
US8383338B2 (en) 2006-04-24 2013-02-26 Roche Nimblegen, Inc. Methods and systems for uniform enrichment of genomic regions
US20070254305A1 (en) 2006-04-28 2007-11-01 Nsabp Foundation, Inc. Methods of whole genome or microarray expression profiling using nucleic acids prepared from formalin fixed paraffin embedded tissue
EP2677039B8 (en) 2006-05-10 2022-10-05 DxTerity Diagnostics Incorporated Detection of nucleic acid targets using chemically reactive oligonucleotide probes
JP2007304043A (en) 2006-05-15 2007-11-22 Canon Inc Method for manufacturing probe-fixing carrier
CA2653095C (en) 2006-05-22 2013-07-16 Nanostring Technologies, Inc. Systems and methods for analyzing nanoreporters
US20080132429A1 (en) 2006-05-23 2008-06-05 Uchicago Argonne Biological microarrays with enhanced signal yield
US9012239B2 (en) 2006-05-27 2015-04-21 Fluidigm Canada Inc. Polymer backbone element tags
US9149564B2 (en) 2006-06-23 2015-10-06 The Regents Of The University Of California Articles comprising large-surface-area bio-compatible materials and methods for making and using them
EP2032686B1 (en) 2006-06-23 2022-01-12 Illumina, Inc. System and method for creation of dna cluster arrays
US8178316B2 (en) 2006-06-29 2012-05-15 President And Fellows Of Harvard College Evaluating proteins
US20090253582A1 (en) 2006-06-29 2009-10-08 Ge Healthcare Bio-Sciences Ab Chamber apparatus
US8362242B2 (en) 2006-06-30 2013-01-29 Dh Technologies Development Pte. Ltd. Analyte determination utilizing mass tagging reagents comprising a non-encoded detectable label
US7312029B1 (en) 2006-06-30 2007-12-25 Searete Llc Method of combing an elongated molecule
AT503862B1 (en) 2006-07-05 2010-11-15 Arc Austrian Res Centers Gmbh PATHOGENIC IDENTIFICATION DUE TO A 16S OR 18S RRNA MICROARRAY
CN101490562B (en) 2006-07-10 2012-12-19 株式会社日立高新技术 Liquid transfer device
EP2081869B1 (en) 2006-07-10 2020-11-04 California Institute of Technology Method for selectively anchoring large numbers of nanoscale structures
EP1878502A1 (en) 2006-07-14 2008-01-16 Roche Diagnostics GmbH Instrument for heating and cooling
CN101522915A (en) 2006-08-02 2009-09-02 加州理工学院 Methods and systems for detecting and/or sorting targets
US9012390B2 (en) 2006-08-07 2015-04-21 Raindance Technologies, Inc. Fluorocarbon emulsion stabilizing surfactants
WO2008022332A2 (en) 2006-08-18 2008-02-21 Board Of Regents, The University Of Texas System System, method and kit for replicating a dna array
US20080047835A1 (en) 2006-08-22 2008-02-28 Macconnell William P Genomic DNA Purifier
EP3936857B1 (en) 2006-09-01 2023-06-21 Pacific Biosciences Of California, Inc. Substrates, systems and methods for analyzing materials
US7754429B2 (en) 2006-10-06 2010-07-13 Illumina Cambridge Limited Method for pair-wise sequencing a plurity of target polynucleotides
EP2076609A1 (en) 2006-10-10 2009-07-08 Illumina Inc. Compositions and methods for representational selection of nucleic acids fro complex mixtures using hybridization
US8343746B2 (en) 2006-10-23 2013-01-01 Pacific Biosciences Of California, Inc. Polymerase enzymes and reagents for enhanced nucleic acid sequencing
US20080108804A1 (en) 2006-11-02 2008-05-08 Kabushiki Kaisha Dnaform Method for modifying RNAS and preparing DNAS from RNAS
US9266076B2 (en) 2006-11-02 2016-02-23 The Regents Of The University Of California Method and apparatus for real-time feedback control of electrical manipulation of droplets on chip
WO2008069906A2 (en) 2006-11-14 2008-06-12 The Regents Of The University Of California Digital expression of gene analysis
US9201063B2 (en) 2006-11-16 2015-12-01 General Electric Company Sequential analysis of biological samples
US7655898B2 (en) 2006-11-30 2010-02-02 Cambridge Research & Instrumentation, Inc. Optical filter assembly with selectable bandwidth and rejection
KR101433208B1 (en) 2006-12-13 2014-08-22 루미넥스 코포레이션 Systems and methods for multiplex analysis of pcr in real time
JP5622392B2 (en) 2006-12-14 2014-11-12 ライフ テクノロジーズ コーポレーション Method and apparatus for analyte measurement using large-scale FET arrays
US8262900B2 (en) 2006-12-14 2012-09-11 Life Technologies Corporation Methods and apparatus for measuring analytes using large scale FET arrays
US8349167B2 (en) 2006-12-14 2013-01-08 Life Technologies Corporation Methods and apparatus for detecting molecular interactions using FET arrays
CN101221182A (en) 2007-01-08 2008-07-16 山东司马特生物芯片有限公司 Novel method for blood serum tumor series diagnosis by fluorescent protein chip
WO2008093098A2 (en) 2007-02-02 2008-08-07 Illumina Cambridge Limited Methods for indexing samples and sequencing multiple nucleotide templates
US20080220434A1 (en) 2007-02-07 2008-09-11 Perscitus Biosciences, Llc Detection Of Molecule Proximity
JP2010517583A (en) 2007-02-12 2010-05-27 プロテオノヴァ、 インコーポレイテッド Generation of a library of soluble random polypeptides linked to mRNA
KR100819006B1 (en) 2007-02-13 2008-04-03 삼성전자주식회사 Mask set for microarray, method of fabricating the same, and method of fabricating microarray using mask set
US8153782B2 (en) 2007-02-14 2012-04-10 Eastman Chemical Company Reformation of ionic liquids
KR101017808B1 (en) 2007-04-04 2011-02-28 엔에이치엔(주) Method for making an edited file automatically and apparatus thereof
JP5555157B2 (en) 2007-04-10 2014-07-23 ナノストリング テクノロジーズ, インコーポレイテッド Method and computer system for identifying target-specific sequences for use in nanoreporters
US20090006002A1 (en) 2007-04-13 2009-01-01 Sequenom, Inc. Comparative sequence analysis processes and systems
WO2008147899A1 (en) 2007-05-23 2008-12-04 Oregon Health & Science University Microarray systems and methods for identifying dna-binding proteins
US20090105959A1 (en) 2007-06-01 2009-04-23 Braverman Michael S System and method for identification of individual samples from a multiplex mixture
WO2008151127A1 (en) 2007-06-04 2008-12-11 President And Fellows Of Harvard College Compounds and methods for chemical ligation
EP2191897B1 (en) 2007-06-21 2014-02-26 Gen-Probe Incorporated Instrument and receptacles for performing processes
CN101679932A (en) 2007-06-27 2010-03-24 数字化生物*** Digital microfluidics based apparatus for heat-exchanging chemical processes
US7635566B2 (en) 2007-06-29 2009-12-22 Population Genetics Technologies Ltd. Methods and compositions for isolating nucleic acid sequence variants
US7534991B2 (en) 2007-07-10 2009-05-19 Cambridge Research & Instrumentation, Inc. Athermalized birefringent filter apparatus and method
US20100112590A1 (en) 2007-07-23 2010-05-06 The Chinese University Of Hong Kong Diagnosing Fetal Chromosomal Aneuploidy Using Genomic Sequencing With Enrichment
US20100159446A1 (en) 2007-07-27 2010-06-24 Haff Lawrence A Detection Assays and Use Thereof
JP2009036694A (en) 2007-08-03 2009-02-19 Tokyo Medical & Dental Univ Method for analyzing biological substance in cell maintaining spatial distribution
WO2009032167A1 (en) 2007-08-29 2009-03-12 Illumina Cambridge Method for sequencing a polynucleotide template
ITBO20070627A1 (en) 2007-09-14 2009-03-15 Twof Inc METHOD FOR THE PREPARATION OF MICROARRAY DNA WITH HIGH LINEAR DENSITY PROBES
US9388457B2 (en) 2007-09-14 2016-07-12 Affymetrix, Inc. Locus specific amplification using array probes
CA2697640C (en) 2007-09-21 2016-06-21 Katholieke Universiteit Leuven Tools and methods for genetic tests using next generation sequencing
US8330087B2 (en) 2007-10-16 2012-12-11 Cambridge Research & Instrumentation, Inc. Spectral imaging system with dynamic optical correction
EP2053132A1 (en) 2007-10-23 2009-04-29 Roche Diagnostics GmbH Enrichment and sequence analysis of geomic regions
US8518640B2 (en) 2007-10-29 2013-08-27 Complete Genomics, Inc. Nucleic acid sequencing and process
US8592150B2 (en) 2007-12-05 2013-11-26 Complete Genomics, Inc. Methods and compositions for long fragment read sequencing
EP3699291A1 (en) 2008-01-17 2020-08-26 Sequenom, Inc. Single molecule nucleic acid sequence analysis processes and compositions
KR20090081260A (en) 2008-01-23 2009-07-28 삼성전자주식회사 Assay method of microarray hybridization
JP5395098B2 (en) 2008-02-15 2014-01-22 バイオ−ラッド ラボラトリーズ,インコーポレイティド Thermal cycler with self-adjusting lid
DE102008014687A1 (en) 2008-03-18 2009-09-24 Smartrac Ip B.V. Layer assembly for a card body and method for producing the layer composite
US7846666B2 (en) 2008-03-21 2010-12-07 Nugen Technologies, Inc. Methods of RNA amplification in the presence of DNA
US20090253163A1 (en) 2008-04-02 2009-10-08 General Electric Company Iterative staining of biological samples
DE102008023438B4 (en) 2008-05-14 2011-06-30 Bruker Daltonik GmbH, 28359 Method for analyzing tissue sections
US8093064B2 (en) 2008-05-15 2012-01-10 The Regents Of The University Of California Method for using magnetic particles in droplet microfluidics
DE102008025656B4 (en) 2008-05-28 2016-07-28 Genxpro Gmbh Method for the quantitative analysis of nucleic acids, markers therefor and their use
WO2009148560A2 (en) 2008-05-30 2009-12-10 Board Of Regents, The Universtiy Of Texas System Methods and compositions for nucleic acid sequencing
US8199999B2 (en) 2008-06-17 2012-06-12 Cambridge Research & Instrumentation, Inc. Image classifier training
GB0811574D0 (en) 2008-06-24 2008-07-30 Trillion Genomics Ltd Characterising planar samples by mass spectrometry
US8198028B2 (en) 2008-07-02 2012-06-12 Illumina Cambridge Limited Using populations of beads for the fabrication of arrays on surfaces
US20100035249A1 (en) 2008-08-05 2010-02-11 Kabushiki Kaisha Dnaform Rna sequencing and analysis using solid support
US8519115B2 (en) 2008-08-14 2013-08-27 Nanostring Technologies, Inc. Stable nanoreporters
EP2326732A4 (en) 2008-08-26 2012-11-14 Fluidigm Corp Assay methods for increased throughput of samples and/or targets
EP2163900A1 (en) 2008-09-04 2010-03-17 Commissariat A L'energie Atomique New method of imaging by mass spectrometry and new mass tag associated trityl derivatives
US20100055733A1 (en) 2008-09-04 2010-03-04 Lutolf Matthias P Manufacture and uses of reactive microcontact printing of biomolecules on soft hydrogels
US8586310B2 (en) 2008-09-05 2013-11-19 Washington University Method for multiplexed nucleic acid patch polymerase chain reaction
US20110244448A1 (en) 2008-09-08 2011-10-06 Masataka Shirai Dna detecting apparatus, dna detecting device and dna detecting method
US8383345B2 (en) 2008-09-12 2013-02-26 University Of Washington Sequence tag directed subassembly of short sequencing reads into long sequencing reads
ES2626903T3 (en) 2008-09-22 2017-07-26 Ventana Medical Systems, Inc. Selective processing of biological material in a microarray substrate
MX2011003380A (en) 2008-09-30 2011-04-21 Abbott Lab Improved antibody libraries.
EP2347252B1 (en) 2008-10-08 2014-07-16 Sage Science, Inc. Multichannel preparative electrophoresis system
US20100137143A1 (en) 2008-10-22 2010-06-03 Ion Torrent Systems Incorporated Methods and apparatus for measuring analytes
US9080211B2 (en) 2008-10-24 2015-07-14 Epicentre Technologies Corporation Transposon end compositions and methods for modifying nucleic acids
US9751084B2 (en) 2008-10-28 2017-09-05 Emd Millipore Corporation Biological culture assembly
EP2356259A4 (en) 2008-10-30 2012-11-14 Sequenom Inc Products and processes for multiplex nucleic acid identification
JP5798926B2 (en) 2008-11-07 2015-10-21 シーケンタ インコーポレイテッド How to monitor disease states by sequence analysis
US8748103B2 (en) 2008-11-07 2014-06-10 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
US9365901B2 (en) 2008-11-07 2016-06-14 Adaptive Biotechnologies Corp. Monitoring immunoglobulin heavy chain evolution in B-cell acute lymphoblastic leukemia
US9394567B2 (en) 2008-11-07 2016-07-19 Adaptive Biotechnologies Corporation Detection and quantification of sample contamination in immune repertoire analysis
EP2370803A2 (en) 2008-12-03 2011-10-05 The United States Government As Represented By The Department Of Veterans Affairs Pressure-assisted molecular recovery (pamr) of biomolecules, pressure-assisted antigen retrieval (paar), and pressure-assisted tissue histology (path)
US20110275077A1 (en) 2009-01-12 2011-11-10 William James Oligonucleotide-Coated Affinity Membranes and Uses Thereof
US8790873B2 (en) 2009-01-16 2014-07-29 Affymetrix, Inc. DNA ligation on RNA template
KR101059565B1 (en) 2009-02-11 2011-08-26 어플라이드 프레시젼, 인코포레이티드 Microarrays with bright reference point labels and methods of collecting optical data therefrom
US8481698B2 (en) 2009-03-19 2013-07-09 The President And Fellows Of Harvard College Parallel proximity ligation event analysis
CA3018687C (en) 2009-04-02 2021-07-13 Fluidigm Corporation Multi-primer amplification method for barcoding of target nucleic acids
CN102625850B (en) 2009-04-03 2014-11-26 蒂莫西·Z·刘 Multiplex nucleic acid detection methods and systems
AU2010242073C1 (en) 2009-04-30 2015-12-24 Good Start Genetics, Inc. Methods and compositions for evaluating genetic markers
WO2010127186A1 (en) 2009-04-30 2010-11-04 Prognosys Biosciences, Inc. Nucleic acid constructs and methods of use
JP5829606B2 (en) 2009-06-29 2015-12-09 カリフォルニア・インスティテュート・オブ・テクノロジーCalifornia Institute Oftechnology Isolation of unknown rearranged T cell receptors from single cells
RU2410439C1 (en) 2009-07-06 2011-01-27 Российская Федерация, От Имени Которой Выступает Министерство Образования И Науки Российской Федерации Method for ablation of target dna from surface of dna biochips
WO2011008831A2 (en) * 2009-07-14 2011-01-20 University Of Florida Research Foundation, Inc. Mass tags for spectrometric analysis of immunoglobulins
GB0912909D0 (en) 2009-07-23 2009-08-26 Olink Genomics Ab Probes for specific analysis of nucleic acids
KR101029343B1 (en) 2009-07-30 2011-04-13 한국과학기술연구원 Immunoassay-based Antigen Detecting Kit and Method
WO2011014811A1 (en) 2009-07-31 2011-02-03 Ibis Biosciences, Inc. Capture primers and capture sequence linked solid supports for molecular diagnostic tests
JP2013500725A (en) 2009-07-31 2013-01-10 プログノシス バイオサイエンシズ インコーポレイテッド Assay tools and methods of use
EP2467479B1 (en) 2009-08-20 2016-01-06 Population Genetics Technologies Ltd Compositions and methods for intramolecular nucleic acid rearrangement
US8496798B2 (en) 2009-09-01 2013-07-30 Oregon Health & Science University Reversible current gel electrophoresis device for separating biological macromolecules
SG169918A1 (en) 2009-10-02 2011-04-29 Fluidigm Corp Microfluidic devices with removable cover and methods of fabrication and application
MX2012004105A (en) 2009-10-09 2012-09-07 Invisible Sentinel Inc Device for detection of antigens and uses thereof.
CA2775613C (en) 2009-10-13 2018-06-12 Nanostring Technologies, Inc. Protein detection via nanoreporters
US9005891B2 (en) 2009-11-10 2015-04-14 Genomic Health, Inc. Methods for depleting RNA from nucleic acid samples
BR112012011181A2 (en) 2009-11-13 2020-10-13 Ventana Medical Systems, Inc. ''automated sliding part processing station and one sample processing method''
US20120277113A1 (en) 2009-11-18 2012-11-01 Ruo-Pan Huang Array-based proximity ligation association assays
CN106701739A (en) 2009-12-04 2017-05-24 株式会社日立制作所 Analysis device and equipment for gene expression analysis
EP2510127B1 (en) 2009-12-07 2015-06-10 Prognosys Biosciences, Inc. Peptide display arrays
SG10201407883PA (en) 2009-12-07 2015-01-29 Illumina Inc Multi-sample indexing for multiplex genotyping
US8835358B2 (en) 2009-12-15 2014-09-16 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse labels
WO2011075083A1 (en) 2009-12-15 2011-06-23 Agency For Science, Technology And Research Processing of amplified dna fragments for sequencing
US8889416B2 (en) 2010-01-21 2014-11-18 California Institute Of Technology Methods and devices for micro-isolation, extraction, and/or analysis of microscale components
WO2011094669A1 (en) 2010-01-29 2011-08-04 Advanced Cell Diagnostics, Inc. Methods of in situ detection of nucleic acids
EP2354242A1 (en) 2010-02-03 2011-08-10 Epiontis GmbH Assay for determining the type and/or status of a cell based on the epigenetic pattern and the chromatin structure
US9714446B2 (en) 2010-02-11 2017-07-25 Nanostring Technologies, Inc. Compositions and methods for the detection of small RNAs
EP2536818B1 (en) 2010-02-18 2018-10-03 Bima Limited Immobilised-bead immunomultiplex assay
JP5665021B2 (en) 2010-03-08 2015-02-04 国立大学法人東京農工大学 Fusion MHC molecule-linked magnetic fine particles, antigen peptide screening method, recombinant vector, and transformant of magnetic bacteria
US10267808B2 (en) 2010-03-08 2019-04-23 California Institute Of Technology Molecular indicia of cellular constituents and resolving the same by super-resolution technologies in single cells
US10266876B2 (en) 2010-03-08 2019-04-23 California Institute Of Technology Multiplex detection of molecular species in cells by super-resolution imaging and combinatorial labeling
US10787701B2 (en) 2010-04-05 2020-09-29 Prognosys Biosciences, Inc. Spatially encoded biological assays
WO2011127099A1 (en) 2010-04-05 2011-10-13 Prognosys Biosciences, Inc. Spatially encoded biological assays
US20110245101A1 (en) 2010-04-05 2011-10-06 Prognosys Biosciences, Inc. Co-localization affinity assays
US8462981B2 (en) 2010-04-07 2013-06-11 Cambridge Research & Instrumentation, Inc. Spectral unmixing for visualization of samples
US10240194B2 (en) 2010-05-13 2019-03-26 Gen9, Inc. Methods for nucleotide sequencing and high fidelity polynucleotide synthesis
US20130059741A1 (en) 2010-05-13 2013-03-07 Illumina, Inc. Binding assays for markers
US8828688B2 (en) 2010-05-27 2014-09-09 Affymetrix, Inc. Multiplex amplification methods
EP2580351B1 (en) 2010-06-09 2018-08-29 Keygene N.V. Combinatorial sequence barcodes for high throughput screening
ES2676183T3 (en) * 2010-07-02 2018-07-17 Ventana Medical Systems, Inc. Target detection using mass marks and mass spectrometry
WO2012012772A2 (en) 2010-07-22 2012-01-26 The Johns Hopkins University Drug eluting hydrogels for catheter delivery
WO2012019133A1 (en) 2010-08-05 2012-02-09 Cambridge Research & Instrumentation, Inc. Enhancing visual assessment of samples
US11203786B2 (en) 2010-08-06 2021-12-21 Ariosa Diagnostics, Inc. Detection of target nucleic acids using hybridization
GB201013767D0 (en) 2010-08-17 2010-09-29 Isis Innovation Identification of ligands and their use
DK2623613T3 (en) 2010-09-21 2016-10-03 Population Genetics Tech Ltd Increasing the reliability of the allele-indications by molecular counting
KR20130113447A (en) 2010-09-24 2013-10-15 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Direct capture, amplification and sequencing of target dna using immobilized primers
US9110079B2 (en) 2010-09-29 2015-08-18 Biomerieux Method and kit for establishing an in vitro prognosis on a patient exhibiting SIRS
WO2012048341A1 (en) 2010-10-08 2012-04-12 President And Fellows Of Harvard College High-throughput single cell barcoding
US10669569B2 (en) 2010-10-15 2020-06-02 Navinci Diagnostics Ab Dynamic range methods
EP3034625B1 (en) 2010-10-21 2017-10-04 Advanced Cell Diagnostics, Inc. An ultra sensitive method for in situ detection of nucleic acids
CA2815076C (en) 2010-10-22 2021-01-12 Cold Spring Harbor Laboratory Varietal counting of nucleic acids for obtaining genomic copy number information
EP2632593B1 (en) 2010-10-27 2021-09-29 Illumina, Inc. Flow cells for biological or chemical analysis
US10024796B2 (en) 2010-10-29 2018-07-17 President And Fellows Of Harvard College Nucleic acid nanostructure barcode probes
WO2012071428A2 (en) 2010-11-22 2012-05-31 Solulink, Inc. Methods and/or use of oligonucleotide conjugates for assays and detections
US20140121118A1 (en) 2010-11-23 2014-05-01 Opx Biotechnologies, Inc. Methods, systems and compositions regarding multiplex construction protein amino-acid substitutions and identification of sequence-activity relationships, to provide gene replacement such as with tagged mutant genes, such as via efficient homologous recombination
EP2652155B1 (en) 2010-12-16 2016-11-16 Gigagen, Inc. Methods for massively parallel analysis of nucleic acids in single cells
US9163281B2 (en) 2010-12-23 2015-10-20 Good Start Genetics, Inc. Methods for maintaining the integrity and identification of a nucleic acid template in a multiplex sequencing reaction
EP3564395A1 (en) 2010-12-30 2019-11-06 Foundation Medicine, Inc. Optimization of multigene analysis of tumor samples
US8951781B2 (en) 2011-01-10 2015-02-10 Illumina, Inc. Systems, methods, and apparatuses to image a sample for biological or chemical analysis
EA201391074A1 (en) 2011-01-25 2013-12-30 Олмак Дайэгностикс Лимитед PROFILES OF EXPRESSION OF GENES OF CANCER OF THICK INTEST. AND METHODS OF APPLICATION
AU2012219132B2 (en) 2011-02-15 2016-05-12 Mats Nilsson Bernitz Method for localized in situ detection of mRNA
EP2689028B1 (en) 2011-03-23 2017-08-30 Pacific Biosciences Of California, Inc. Isolation of polymerase-nucleic acid complexes and loading onto substrates
US9260753B2 (en) 2011-03-24 2016-02-16 President And Fellows Of Harvard College Single cell nucleic acid detection and analysis
US20120258871A1 (en) 2011-04-08 2012-10-11 Prognosys Biosciences, Inc. Peptide constructs and assay systems
GB201106254D0 (en) 2011-04-13 2011-05-25 Frisen Jonas Method and product
AU2012242847B2 (en) 2011-04-15 2017-01-19 The Johns Hopkins University Safe sequencing system
US8946389B2 (en) 2011-04-25 2015-02-03 University Of Washington Compositions and methods for multiplex biomarker profiling
CA2834976C (en) 2011-05-04 2016-03-15 Htg Molecular Diagnostics, Inc. Quantitative nuclease protection assay (qnpa) and sequencing (qnps) improvements
SG194722A1 (en) 2011-05-09 2013-12-30 Fluidigm Corp Probe based nucleic acid detection
DK2710145T3 (en) 2011-05-17 2016-02-22 Dxterity Diagnostics Inc METHOD AND COMPOSITIONS FOR THE DETECTION OF TARGET NUCLEIC ACIDS
WO2012159089A1 (en) 2011-05-19 2012-11-22 Sequenom, Inc. Products and processes for multiplex nucleic acid identification
US9005935B2 (en) 2011-05-23 2015-04-14 Agilent Technologies, Inc. Methods and compositions for DNA fragmentation and tagging by transposases
GB201108678D0 (en) 2011-05-24 2011-07-06 Olink Ab Multiplexed proximity ligation assay
WO2012168003A1 (en) 2011-06-06 2012-12-13 Biocartis S.A. Selective lysis of cells by ionic surfactants
JP6121409B2 (en) 2011-06-17 2017-04-26 ロッシュ ダイアグノスティクス ヘマトロジー インコーポレイテッド Solutions for tissue processing of biological samples
WO2013019960A1 (en) 2011-08-03 2013-02-07 Bio-Rad Laboratories, Inc. Filtering small nucleic acids using permeabilized cells
WO2013033271A2 (en) 2011-08-29 2013-03-07 Derren Barken Method to augment immune system in response to disease or injury
US10385475B2 (en) 2011-09-12 2019-08-20 Adaptive Biotechnologies Corp. Random array sequencing of low-complexity libraries
SI3623481T1 (en) 2011-09-23 2022-01-31 Illumina, Inc. Compositions for nucleic acid sequencing
US10501791B2 (en) 2011-10-14 2019-12-10 President And Fellows Of Harvard College Sequencing by structure assembly
EP2771103B1 (en) 2011-10-28 2017-08-16 Illumina, Inc. Microarray fabrication system and method
US8987174B2 (en) 2011-10-28 2015-03-24 Prognosys Biosciences, Inc. Methods for manufacturing molecular arrays
CA2854023A1 (en) 2011-11-07 2013-05-16 Illumina, Inc. Integrated sequencing apparatuses and methods of use
DK2788499T3 (en) 2011-12-09 2016-03-21 Illumina Inc Enhanced root for polymer tags
US20150031553A1 (en) 2011-12-13 2015-01-29 Sequenta, Inc. Method of measuring immune activation
US9803188B2 (en) 2011-12-22 2017-10-31 Ibis Biosciences, Inc. Systems and methods for isolating nucleic acids
US11021737B2 (en) 2011-12-22 2021-06-01 President And Fellows Of Harvard College Compositions and methods for analyte detection
ES2953308T3 (en) 2011-12-22 2023-11-10 Harvard College Compositions and methods for the detection of analytes
EP2814986B1 (en) 2012-02-14 2017-10-25 Cornell University Method for relative quantification of nucleic acid sequence, expression, or copy changes, using combined nuclease, ligation, and polymerase reactions
US10011871B2 (en) 2012-02-17 2018-07-03 Fred Hutchinson Cancer Research Center Compositions and methods for accurately identifying mutations
NO2694769T3 (en) 2012-03-06 2018-03-03
SG11201405669XA (en) 2012-03-13 2014-10-30 Swift Biosciences Inc Methods and compositions for size-controlled homopolymer tailing of substrate polynucleotides by a nucleic acid polymerase
CA2867293C (en) 2012-03-13 2020-09-01 Abhijit Ajit PATEL Measurement of nucleic acid variants using highly-multiplexed error-suppressed deep sequencing
ES2828661T3 (en) 2012-03-20 2021-05-27 Univ Washington Through Its Center For Commercialization Methods to Reduce the Error Rate of Parallel Massive DNA Sequencing Using Double-stranded Consensus Sequence Sequencing
FI2831279T3 (en) 2012-03-26 2023-05-23 Univ Johns Hopkins Rapid aneuploidy detection
EP2647426A1 (en) 2012-04-03 2013-10-09 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Replication of distributed nucleic acid molecules with preservation of their relative distribution through hybridization-based binding
US9193996B2 (en) 2012-04-03 2015-11-24 Illumina, Inc. Integrated optoelectronic read head and fluidic cartridge useful for nucleic acid sequencing
EP2659977B1 (en) 2012-05-02 2019-04-24 IMEC vzw Microfluidics system for sequencing
US9012022B2 (en) 2012-06-08 2015-04-21 Illumina, Inc. Polymer coatings
US8895249B2 (en) 2012-06-15 2014-11-25 Illumina, Inc. Kinetic exclusion amplification of nucleic acid libraries
AU2013302756C1 (en) 2012-08-14 2018-05-17 10X Genomics, Inc. Microcapsule compositions and methods
US20140378345A1 (en) 2012-08-14 2014-12-25 10X Technologies, Inc. Compositions and methods for sample processing
ES2660027T3 (en) 2012-10-01 2018-03-20 Adaptive Biotechnologies Corporation Evaluation of immunocompetence by the diversity of adaptive immunity receptors and clonal characterization
EP3511423B1 (en) 2012-10-17 2021-03-24 Spatial Transcriptomics AB Methods and product for optimising localised or spatial detection of gene expression in a tissue sample
EP2722105A1 (en) 2012-10-22 2014-04-23 Universität Wien Method of in situ synthesizing microarrays
CN117845337A (en) 2012-12-10 2024-04-09 分析生物科学有限公司 Methods of targeted genomic analysis
US9512422B2 (en) 2013-02-26 2016-12-06 Illumina, Inc. Gel patterned surfaces
WO2014200767A1 (en) 2013-06-12 2014-12-18 The General Hospital Corporation Methods, kits, and systems for multiplexed detection of target molecules and uses thereof
WO2014210225A1 (en) 2013-06-25 2014-12-31 Prognosys Biosciences, Inc. Methods and systems for determining spatial patterns of biological targets in a sample
DE202015009494U1 (en) 2014-04-10 2018-02-08 10X Genomics, Inc. Fluidic devices and systems for encapsulating and partitioning reagents, and their applications
CN107002128A (en) 2014-10-29 2017-08-01 10X 基因组学有限公司 The method and composition being sequenced for target nucleic acid
EP3234192B1 (en) 2014-12-19 2021-07-14 The Broad Institute, Inc. Unbiased identification of double-strand breaks and genomic rearrangement by genome-wide insert capture sequencing
EP3530752B1 (en) 2015-04-10 2021-03-24 Spatial Transcriptomics AB Spatially distinguished, multiplex nucleic acid analysis of biological specimens
US10059990B2 (en) 2015-04-14 2018-08-28 Massachusetts Institute Of Technology In situ nucleic acid sequencing of expanded biological samples
US10633648B2 (en) 2016-02-12 2020-04-28 University Of Washington Combinatorial photo-controlled spatial sequencing and labeling
US20170253918A1 (en) 2016-03-01 2017-09-07 Expansion Technologies Combining protein barcoding with expansion microscopy for in-situ, spatially-resolved proteomics
KR20230047506A (en) 2016-05-02 2023-04-07 엔코디아, 인코포레이티드 Macromolecule analysis employing nucleic acid encoding
US20180052081A1 (en) 2016-05-11 2018-02-22 Expansion Technologies Combining modified antibodies with expansion microscopy for in-situ, spatially-resolved proteomics
CN114875125A (en) 2016-10-19 2022-08-09 10X基因组学有限公司 Methods and systems for barcoding nucleic acid molecules of individual cells or cell populations
US20190177800A1 (en) 2017-12-08 2019-06-13 10X Genomics, Inc. Methods and compositions for labeling cells
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
EP4029939B1 (en) 2017-01-30 2023-06-28 10X Genomics, Inc. Methods and systems for droplet-based single cell barcoding
US20180312822A1 (en) 2017-04-26 2018-11-01 10X Genomics, Inc. Mmlv reverse transcriptase variants
US10549279B2 (en) 2017-08-22 2020-02-04 10X Genomics, Inc. Devices having a plurality of droplet formation regions
WO2019157529A1 (en) 2018-02-12 2019-08-15 10X Genomics, Inc. Methods characterizing multiple analytes from individual cells or cell populations
EP3775271A1 (en) 2018-04-06 2021-02-17 10X Genomics, Inc. Systems and methods for quality control in single cell processing
US11519033B2 (en) 2018-08-28 2022-12-06 10X Genomics, Inc. Method for transposase-mediated spatial tagging and analyzing genomic DNA in a biological sample
US20210324457A1 (en) 2018-08-28 2021-10-21 Eswar Prasad Ramachandran Iyer Methods for Generating Spatially Barcoded Arrays
SG11202102029TA (en) 2018-08-28 2021-03-30 10X Genomics Inc Methods for generating spatially barcoded arrays
US20210317524A1 (en) 2018-08-28 2021-10-14 10X Genomics, Inc. Resolving spatial arrays
WO2020076979A1 (en) 2018-10-10 2020-04-16 Readcoor, Inc. Surface capture of targets
EP3894587A1 (en) 2018-12-10 2021-10-20 10X Genomics, Inc. Resolving spatial arrays by proximity-based deconvolution
US20210189475A1 (en) 2018-12-10 2021-06-24 10X Genomics, Inc. Imaging system hardware
US20220049293A1 (en) 2018-12-10 2022-02-17 10X Genomics, Inc. Methods for determining a location of a biological analyte in a biological sample
US20220267844A1 (en) 2019-11-27 2022-08-25 10X Genomics, Inc. Methods for determining a location of a biological analyte in a biological sample
US11649485B2 (en) 2019-01-06 2023-05-16 10X Genomics, Inc. Generating capture probes for spatial analysis
WO2020167862A1 (en) 2019-02-12 2020-08-20 10X Genomics, Inc. Systems and methods for transfer of reagents between droplets
WO2020176882A1 (en) 2019-02-28 2020-09-03 10X Genomics, Inc. Devices, systems, and methods for increasing droplet formation efficiency
US20230159995A1 (en) 2019-02-28 2023-05-25 10X Genomics, Inc. Profiling of biological analytes with spatially barcoded oligonucleotide arrays
EP3931354A1 (en) 2019-02-28 2022-01-05 10X Genomics, Inc. Profiling of biological analytes with spatially barcoded oligonucleotide arrays
US20220145361A1 (en) 2019-03-15 2022-05-12 10X Genomics, Inc. Methods for using spatial arrays for single cell sequencing
EP3938538A1 (en) 2019-03-15 2022-01-19 10X Genomics, Inc. Methods for using spatial arrays for single cell sequencing
WO2020198071A1 (en) 2019-03-22 2020-10-01 10X Genomics, Inc. Three-dimensional spatial analysis
US20220017951A1 (en) 2019-03-22 2022-01-20 10X Genomics, Inc. Three-dimensional spatial analysis
WO2020219901A1 (en) 2019-04-26 2020-10-29 10X Genomics, Inc. Imaging support devices
WO2020243579A1 (en) 2019-05-30 2020-12-03 10X Genomics, Inc. Methods of detecting spatial heterogeneity of a biological sample
US20210140982A1 (en) 2019-10-18 2021-05-13 10X Genomics, Inc. Identification of spatial biomarkers of brain disorders and methods of using the same
WO2021091611A1 (en) 2019-11-08 2021-05-14 10X Genomics, Inc. Spatially-tagged analyte capture agents for analyte multiplexing
EP4025711A2 (en) 2019-11-08 2022-07-13 10X Genomics, Inc. Enhancing specificity of analyte binding
US20230002812A1 (en) 2019-11-13 2023-01-05 10X Genomics, Inc. Generating capture probes for spatial analysis
US20210199660A1 (en) 2019-11-22 2021-07-01 10X Genomics, Inc. Biomarkers of breast cancer
WO2021133842A1 (en) 2019-12-23 2021-07-01 10X Genomics, Inc. Compositions and methods for using fixed biological samples in partition-based assays
EP3891300B1 (en) 2019-12-23 2023-03-29 10X Genomics, Inc. Methods for spatial analysis using rna-templated ligation
WO2021133845A1 (en) 2019-12-23 2021-07-01 10X Genomics, Inc. Reversible fixing reagents and methods of use thereof
US20210198741A1 (en) 2019-12-30 2021-07-01 10X Genomics, Inc. Identification of spatial biomarkers of heart disorders and methods of using the same
US20220348992A1 (en) 2020-01-10 2022-11-03 10X Genomics, Inc. Methods for determining a location of a target nucleic acid in a biological sample
CN115715329A (en) 2020-01-10 2023-02-24 10X基因组学有限公司 Method for determining the position of a target nucleic acid in a biological sample
US20210214785A1 (en) 2020-01-13 2021-07-15 Spatial Transcriptomics Ab Methods of decreasing background on a spatial array
US20210223227A1 (en) 2020-01-17 2021-07-22 Spatial Transcriptomics Ab Electrophoretic system and method for analyte capture
US11732299B2 (en) 2020-01-21 2023-08-22 10X Genomics, Inc. Spatial assays with perturbed cells
US11702693B2 (en) 2020-01-21 2023-07-18 10X Genomics, Inc. Methods for printing cells and generating arrays of barcoded cells
US20210222253A1 (en) 2020-01-21 2021-07-22 10X Genomics, Inc. Identification of biomarkers of glioblastoma and methods of using the same
US20210230681A1 (en) 2020-01-24 2021-07-29 10X Genomics, Inc. Methods for spatial analysis using proximity ligation
US20210237022A1 (en) 2020-01-31 2021-08-05 10X Genomics, Inc. Capturing oligonucleotides in spatial transcriptomics
US20210238664A1 (en) 2020-02-03 2021-08-05 10X Genomics, Inc. Methods for preparing high-resolution spatial arrays
US11898205B2 (en) 2020-02-03 2024-02-13 10X Genomics, Inc. Increasing capture efficiency of spatial assays
US11732300B2 (en) 2020-02-05 2023-08-22 10X Genomics, Inc. Increasing efficiency of spatial analysis in a biological sample
US20230047782A1 (en) 2020-02-07 2023-02-16 10X Genomics, Inc. Quantitative and automated permeabilization performance evaluation for spatial transcriptomics
US11835462B2 (en) 2020-02-11 2023-12-05 10X Genomics, Inc. Methods and compositions for partitioning a biological sample
WO2021168278A1 (en) 2020-02-20 2021-08-26 10X Genomics, Inc. METHODS TO COMBINE FIRST AND SECOND STRAND cDNA SYNTHESIS FOR SPATIAL ANALYSIS
AU2021224760A1 (en) 2020-02-21 2022-09-15 10X Genomics, Inc. Capturing genetic targets using a hybridization approach
US11891654B2 (en) 2020-02-24 2024-02-06 10X Genomics, Inc. Methods of making gene expression libraries
WO2021207610A1 (en) 2020-04-10 2021-10-14 10X Genomics, Inc. Cold protease treatment method for preparing biological samples
WO2021216708A1 (en) 2020-04-22 2021-10-28 10X Genomics, Inc. Methods for spatial analysis using targeted rna depletion
EP4146819A1 (en) 2020-05-04 2023-03-15 10X Genomics, Inc. Spatial transcriptomic transfer modes
US20230194469A1 (en) 2020-05-19 2023-06-22 10X Genomics, Inc. Electrophoresis cassettes and instrumentation
WO2021236929A1 (en) 2020-05-22 2021-11-25 10X Genomics, Inc. Simultaneous spatio-temporal measurement of gene expression and cellular activity
WO2021237056A1 (en) 2020-05-22 2021-11-25 10X Genomics, Inc. Rna integrity analysis in a biological sample
WO2021237087A1 (en) 2020-05-22 2021-11-25 10X Genomics, Inc. Spatial analysis to detect sequence variants
WO2021242834A1 (en) 2020-05-26 2021-12-02 10X Genomics, Inc. Method for resetting an array
AU2021283174A1 (en) 2020-06-02 2023-01-05 10X Genomics, Inc. Nucleic acid library methods
WO2021247568A1 (en) 2020-06-02 2021-12-09 10X Genomics, Inc. Spatial trancriptomics for antigen-receptors
EP4162074B1 (en) 2020-06-08 2024-04-24 10X Genomics, Inc. Methods of determining a surgical margin and methods of use thereof
WO2021252591A1 (en) 2020-06-10 2021-12-16 10X Genomics, Inc. Methods for determining a location of an analyte in a biological sample
US20230279474A1 (en) 2020-06-10 2023-09-07 10X Genomics, Inc. Methods for spatial analysis using blocker oligonucleotides
CN116034166A (en) 2020-06-25 2023-04-28 10X基因组学有限公司 Spatial analysis of DNA methylation
CN116323968A (en) 2020-07-31 2023-06-23 10X基因组学有限公司 Decrosslinking compounds for spatial analysis and methods of use
EP4200441A1 (en) 2020-09-15 2023-06-28 10X Genomics, Inc. Methods of releasing an extended capture probe from a substrate and uses of the same
AU2021345133A1 (en) 2020-09-16 2023-03-30 10X Genomics, Inc. Methods of determining the location of an analyte in a biological sample using a plurality of wells
WO2022087273A1 (en) 2020-10-22 2022-04-28 10X Genomics, Inc. Methods for spatial analysis using rolling circle amplification
EP4222283A1 (en) 2020-11-06 2023-08-09 10X Genomics, Inc. Compositions and methods for binding an analyte to a capture probe
WO2022098810A1 (en) 2020-11-06 2022-05-12 10X Genomics, Inc. Assay support devices
WO2022103712A1 (en) 2020-11-13 2022-05-19 10X Genomics, Inc. Nano-partitions (encapsulated nucleic acid processing enzymes) for cell-lysis and multiple reactions in partition-based assays
AU2021385065A1 (en) 2020-11-18 2023-06-15 10X Genomics, Inc. Methods and compositions for analyzing immune infiltration in cancer stroma to predict clinical outcome
AU2021409136A1 (en) 2020-12-21 2023-06-29 10X Genomics, Inc. Methods, compositions, and systems for capturing probes and/or barcodes
WO2022147296A1 (en) 2020-12-30 2022-07-07 10X Genomics, Inc. Cleavage of capture probes for spatial analysis
US20240068017A1 (en) 2020-12-30 2024-02-29 10X Genomics, Inc. Methods for analyte capture determination
US20240093290A1 (en) 2021-01-29 2024-03-21 10X Genomics, Inc. Method for transposase mediated spatial tagging and analyzing genomic dna in a biological sample
EP4294571A2 (en) 2021-02-19 2023-12-27 10X Genomics, Inc. Modular assay support devices
EP4301870A1 (en) 2021-03-18 2024-01-10 10X Genomics, Inc. Multiplex capture of gene and protein expression from a biological sample
EP4305196A1 (en) 2021-04-14 2024-01-17 10X Genomics, Inc. Methods of measuring mislocalization of an analyte
WO2022226057A1 (en) 2021-04-20 2022-10-27 10X Genomics, Inc. Methods for assessing sample quality prior to spatial analysis using templated ligation
US20220333192A1 (en) 2021-04-20 2022-10-20 10X Genomics, Inc. Methods and devices for spatial assessment of rna quality
EP4320271A1 (en) 2021-05-06 2024-02-14 10X Genomics, Inc. Methods for increasing resolution of spatial analysis
WO2022256503A1 (en) 2021-06-03 2022-12-08 10X Genomics, Inc. Methods, compositions, kits, and systems for enhancing analyte capture for spatial analysis
WO2022271820A1 (en) 2021-06-22 2022-12-29 10X Genomics, Inc. Spatial detection of sars-cov-2 using templated ligation
US20230014008A1 (en) 2021-07-13 2023-01-19 10X Genomics, Inc. Methods for improving spatial performance
WO2023287765A1 (en) 2021-07-13 2023-01-19 10X Genomics, Inc. Methods for spatial analysis using targeted probe silencing
US20230034216A1 (en) 2021-07-28 2023-02-02 10X Genomics, Inc. Multiplexed spatial capture of analytes
US20230034039A1 (en) 2021-08-02 2023-02-02 10X Genomics, Inc. Methods of preserving a biological sample
US20230042817A1 (en) 2021-08-04 2023-02-09 10X Genomics, Inc. Analyte capture from an embedded biological sample
WO2023018799A1 (en) 2021-08-12 2023-02-16 10X Genomics, Inc. Methods, compositions and systems for identifying antigen-binding molecules
EP4196605A1 (en) 2021-09-01 2023-06-21 10X Genomics, Inc. Methods, compositions, and kits for blocking a capture probe on a spatial array
US20230135010A1 (en) 2021-11-03 2023-05-04 10X Genomics, Inc. Sequential analyte capture
WO2023086880A1 (en) 2021-11-10 2023-05-19 10X Genomics, Inc. Methods, compositions, and kits for determining the location of an analyte in a biological sample

Cited By (68)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11795498B2 (en) 2011-04-13 2023-10-24 10X Genomics Sweden Ab Methods of detecting analytes
US11352659B2 (en) 2011-04-13 2022-06-07 Spatial Transcriptomics Ab Methods of detecting analytes
US11788122B2 (en) 2011-04-13 2023-10-17 10X Genomics Sweden Ab Methods of detecting analytes
US11479809B2 (en) 2011-04-13 2022-10-25 Spatial Transcriptomics Ab Methods of detecting analytes
US11618918B2 (en) 2013-06-25 2023-04-04 Prognosys Biosciences, Inc. Methods and systems for determining spatial patterns of biological targets in a sample
US11286515B2 (en) 2013-06-25 2022-03-29 Prognosys Biosciences, Inc. Methods and systems for determining spatial patterns of biological targets in a sample
US11821024B2 (en) 2013-06-25 2023-11-21 Prognosys Biosciences, Inc. Methods and systems for determining spatial patterns of biological targets in a sample
US11359228B2 (en) 2013-06-25 2022-06-14 Prognosys Biosciences, Inc. Methods and systems for determining spatial patterns of biological targets in a sample
US11753674B2 (en) 2013-06-25 2023-09-12 Prognosys Biosciences, Inc. Methods and systems for determining spatial patterns of biological targets in a sample
US11162132B2 (en) 2015-04-10 2021-11-02 Spatial Transcriptomics Ab Spatially distinguished, multiplex nucleic acid analysis of biological specimens
US11739372B2 (en) 2015-04-10 2023-08-29 Spatial Transcriptomics Ab Spatially distinguished, multiplex nucleic acid analysis of biological specimens
US11390912B2 (en) 2015-04-10 2022-07-19 Spatial Transcriptomics Ab Spatially distinguished, multiplex nucleic acid analysis of biological specimens
US11613773B2 (en) 2015-04-10 2023-03-28 Spatial Transcriptomics Ab Spatially distinguished, multiplex nucleic acid analysis of biological specimens
US11299774B2 (en) 2015-04-10 2022-04-12 Spatial Transcriptomics Ab Spatially distinguished, multiplex nucleic acid analysis of biological specimens
US11519033B2 (en) 2018-08-28 2022-12-06 10X Genomics, Inc. Method for transposase-mediated spatial tagging and analyzing genomic DNA in a biological sample
US11933957B1 (en) 2018-12-10 2024-03-19 10X Genomics, Inc. Imaging system hardware
US11926867B2 (en) 2019-01-06 2024-03-12 10X Genomics, Inc. Generating capture probes for spatial analysis
US11649485B2 (en) 2019-01-06 2023-05-16 10X Genomics, Inc. Generating capture probes for spatial analysis
US11753675B2 (en) 2019-01-06 2023-09-12 10X Genomics, Inc. Generating capture probes for spatial analysis
US11965213B2 (en) 2019-05-30 2024-04-23 10X Genomics, Inc. Methods of detecting spatial heterogeneity of a biological sample
US11702698B2 (en) 2019-11-08 2023-07-18 10X Genomics, Inc. Enhancing specificity of analyte binding
US11592447B2 (en) 2019-11-08 2023-02-28 10X Genomics, Inc. Spatially-tagged analyte capture agents for analyte multiplexing
US11808769B2 (en) 2019-11-08 2023-11-07 10X Genomics, Inc. Spatially-tagged analyte capture agents for analyte multiplexing
US11795507B2 (en) 2019-12-23 2023-10-24 10X Genomics, Inc. Methods for spatial analysis using RNA-templated ligation
US11505828B2 (en) 2019-12-23 2022-11-22 10X Genomics, Inc. Methods for spatial analysis using RNA-templated ligation
US11560593B2 (en) 2019-12-23 2023-01-24 10X Genomics, Inc. Methods for spatial analysis using RNA-templated ligation
US11332790B2 (en) 2019-12-23 2022-05-17 10X Genomics, Inc. Methods for spatial analysis using RNA-templated ligation
US11732299B2 (en) 2020-01-21 2023-08-22 10X Genomics, Inc. Spatial assays with perturbed cells
US11702693B2 (en) 2020-01-21 2023-07-18 10X Genomics, Inc. Methods for printing cells and generating arrays of barcoded cells
US11821035B1 (en) 2020-01-29 2023-11-21 10X Genomics, Inc. Compositions and methods of making gene expression libraries
US11898205B2 (en) 2020-02-03 2024-02-13 10X Genomics, Inc. Increasing capture efficiency of spatial assays
US11732300B2 (en) 2020-02-05 2023-08-22 10X Genomics, Inc. Increasing efficiency of spatial analysis in a biological sample
US11835462B2 (en) 2020-02-11 2023-12-05 10X Genomics, Inc. Methods and compositions for partitioning a biological sample
US11891654B2 (en) 2020-02-24 2024-02-06 10X Genomics, Inc. Methods of making gene expression libraries
US11926863B1 (en) 2020-02-27 2024-03-12 10X Genomics, Inc. Solid state single cell method for analyzing fixed biological cells
US11768175B1 (en) 2020-03-04 2023-09-26 10X Genomics, Inc. Electrophoretic methods for spatial analysis
US11773433B2 (en) 2020-04-22 2023-10-03 10X Genomics, Inc. Methods for spatial analysis using targeted RNA depletion
US11535887B2 (en) 2020-04-22 2022-12-27 10X Genomics, Inc. Methods for spatial analysis using targeted RNA depletion
US11608520B2 (en) 2020-05-22 2023-03-21 10X Genomics, Inc. Spatial analysis to detect sequence variants
US11959130B2 (en) 2020-05-22 2024-04-16 10X Genomics, Inc. Spatial analysis to detect sequence variants
US11866767B2 (en) 2020-05-22 2024-01-09 10X Genomics, Inc. Simultaneous spatio-temporal measurement of gene expression and cellular activity
US11624086B2 (en) 2020-05-22 2023-04-11 10X Genomics, Inc. Simultaneous spatio-temporal measurement of gene expression and cellular activity
US11560592B2 (en) 2020-05-26 2023-01-24 10X Genomics, Inc. Method for resetting an array
US11692218B2 (en) 2020-06-02 2023-07-04 10X Genomics, Inc. Spatial transcriptomics for antigen-receptors
US11840687B2 (en) 2020-06-02 2023-12-12 10X Genomics, Inc. Nucleic acid library methods
US11512308B2 (en) 2020-06-02 2022-11-29 10X Genomics, Inc. Nucleic acid library methods
US11608498B2 (en) 2020-06-02 2023-03-21 10X Genomics, Inc. Nucleic acid library methods
US11845979B2 (en) 2020-06-02 2023-12-19 10X Genomics, Inc. Spatial transcriptomics for antigen-receptors
US11859178B2 (en) 2020-06-02 2024-01-02 10X Genomics, Inc. Nucleic acid library methods
US11492612B1 (en) 2020-06-08 2022-11-08 10X Genomics, Inc. Methods of determining a surgical margin and methods of use thereof
US11407992B2 (en) 2020-06-08 2022-08-09 10X Genomics, Inc. Methods of determining a surgical margin and methods of use thereof
US11781130B2 (en) 2020-06-08 2023-10-10 10X Genomics, Inc. Methods of determining a surgical margin and methods of use thereof
US11624063B2 (en) 2020-06-08 2023-04-11 10X Genomics, Inc. Methods of determining a surgical margin and methods of use thereof
US11434524B2 (en) 2020-06-10 2022-09-06 10X Genomics, Inc. Methods for determining a location of an analyte in a biological sample
US11408029B2 (en) 2020-06-25 2022-08-09 10X Genomics, Inc. Spatial analysis of DNA methylation
US11661626B2 (en) 2020-06-25 2023-05-30 10X Genomics, Inc. Spatial analysis of DNA methylation
US11761038B1 (en) 2020-07-06 2023-09-19 10X Genomics, Inc. Methods for identifying a location of an RNA in a biological sample
US11952627B2 (en) 2020-07-06 2024-04-09 10X Genomics, Inc. Methods for identifying a location of an RNA in a biological sample
US11926822B1 (en) 2020-09-23 2024-03-12 10X Genomics, Inc. Three-dimensional spatial analysis
US11827935B1 (en) 2020-11-19 2023-11-28 10X Genomics, Inc. Methods for spatial analysis using rolling circle amplification and detection probes
US11680260B2 (en) 2020-12-21 2023-06-20 10X Genomics, Inc. Methods, compositions, and systems for spatial analysis of analytes in a biological sample
US11873482B2 (en) 2020-12-21 2024-01-16 10X Genomics, Inc. Methods, compositions, and systems for spatial analysis of analytes in a biological sample
US11618897B2 (en) 2020-12-21 2023-04-04 10X Genomics, Inc. Methods, compositions, and systems for capturing probes and/or barcodes
US11959076B2 (en) 2020-12-21 2024-04-16 10X Genomics, Inc. Methods, compositions, and systems for capturing probes and/or barcodes
US11739381B2 (en) 2021-03-18 2023-08-29 10X Genomics, Inc. Multiplex capture of gene and protein expression from a biological sample
US11753673B2 (en) 2021-09-01 2023-09-12 10X Genomics, Inc. Methods, compositions, and kits for blocking a capture probe on a spatial array
US11840724B2 (en) 2021-09-01 2023-12-12 10X Genomics, Inc. Methods, compositions, and kits for blocking a capture probe on a spatial array
US11970739B2 (en) 2023-07-06 2024-04-30 10X Genomics, Inc. Multiplex capture of gene and protein expression from a biological sample

Also Published As

Publication number Publication date
US11733238B2 (en) 2023-08-22
US20200325531A1 (en) 2020-10-15

Similar Documents

Publication Publication Date Title
US11519022B2 (en) Spatially encoded biological assays
US11156603B2 (en) Spatially encoded biological assays
US20190300945A1 (en) Spatially Encoded Biological Assays

Legal Events

Date Code Title Description
AS Assignment

Owner name: PROGNOSYS BIOSCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHEE, MARK S.;REEL/FRAME:049440/0778

Effective date: 20130717

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION