Selective amplification using blocking oligonucleotides

Information

  • Patent Grant
  • 11319583
  • Patent Number
    11,319,583
  • Date Filed
    Friday, January 19, 2018
    6 years ago
  • Date Issued
    Tuesday, May 3, 2022
    2 years ago
Abstract
Disclosed herein include methods and compositions for selectively amplifying and/or extending nucleic acid target molecules in a sample. The methods and compositions can, for example, reduce the amplification and/or extension of undesirable nucleic acid species in the sample, and/or allow selective removal of undesirable nucleic acid species in the sample.
Description
BACKGROUND

The expression level of different genes can vary significantly in a biological sample. For examples, some broad categories of gene expression are: 1) “high expressers” which are comprised of 5-10 genes that dominate ˜20% of cellular mRNAs; 2) “intermediate expressers” that are comprised of 50-200 genes that occupy 40-60% of cellular mRNAs; and 3) “moderate expressers” that are comprised of 10,000-20,000 genes that occupy the rest of the cellular mRNA fraction. One challenge in molecular biology and molecular genetics is to capture this highly dynamic gene expression profile efficiently and accurately in order to distinguish different cell types and phenotypes in the sample.


In recent years, next generation sequencing (NGS) has provided a high throughput method in assessing gene expression profiles. During library preparation for NGS, a sample with heterogeneous cDNA species is amplified by PCR to obtain adequate sample amount and to attach NGS-compatible adapters. The sequencing process captures the number of reads for each gene from the PCR-amplified library sample to interpret the gene expression level. However, since different genes are expressed at a large range of levels, PCR amplification can skew the native gene expression. For example, a gene has 1 molecule of cDNA would require 40 cycles of PCR to achieve the same representative amount as a gene with 1000 molecules of cDNA in 30 cycles. In a heterogeneous cDNA sample, PCR is usually performed in excess cycles to adequately amplify low expressers; in those scenarios, the native gene expression profile is usually skewed by the dominating high expresser PCR products. A method to correct for such a bias in PCR product is Molecular Indexing; however, high expressers such as ribosomal protein mRNAs, mitochondrial mRNAs, or housekeeping genes often dominate the sequencing run with little contribution to the experimental interpretation. There is a need for selectively amplifying sequences of interest.


SUMMARY

Disclosed herein is a method of selective amplification. In some embodiments, the method comprises: providing a sample comprising a plurality of nucleic acid target molecules and one or more undesirable nucleic acid species; providing a plurality of oligonucleotide probes, wherein each of the plurality of oligonucleotide probes comprises a molecular label sequence and a binding region; contacting the plurality of oligonucleotide probes with the plurality of nucleic acid target molecules for hybridization; extending oligonucleotide probes that are hybridized to the plurality of nucleic acid target molecules to generate a plurality of extension products; providing a blocking oligonucleotide that specifically binds to at least one of the one or more undesirable nucleic acid species; and amplifying the plurality of extension products to generate a plurality of amplicons, whereby the amplification or the extension of the undesirable nucleic acid species is reduced by the blocking oligonucleotide. Also disclosed herein is a method of selective extension. In some embodiments, the method comprises: providing a sample comprising a plurality of nucleic acid target molecules and one or more undesirable nucleic acid species; providing a plurality of oligonucleotide probes, wherein each of the plurality of oligonucleotide probes comprises a molecular label sequence and a binding region; contacting the plurality of oligonucleotide probes with the plurality of nucleic acid target molecules for hybridization; providing a blocking oligonucleotide that specifically binds to at least one of the one or more undesirable nucleic acid species; and extending oligonucleotide probes that are hybridized to the plurality of nucleic acid target molecules to generate a plurality of extension products; whereby the extension of the undesirable nucleic acid species is reduced by the blocking oligonucleotide.


In the methods and compositions disclosed herein, the blocking oligonucleotide can be, for example, a locked nucleic acid (LNA), a peptide nucleic acid (PNA), a DNA, an LNA/PNA chimera, an LNA/DNA chimera, or a PNA/DNA chimera. In some embodiments, the methods comprise providing blocking oligonucleotides that specifically binds to two or more undesirable nucleic acid species in the sample. In some embodiments, the methods comprise providing blocking oligonucleotides that specifically binds to at least 10 undesirable nucleic acid species in the sample. In some embodiments, the methods comprise providing blocking oligonucleotides that specifically binds to at least 100 undesirable nucleic acid species in the sample.


The blocking oligonucleotide can have a Tm of at least 60° C., a Tm of at least 65° C., or a Tm of at least 70° C. In some embodiments, the blocking oligonucleotide is unable to function as a primer for a reverse transcriptase or a polymerase. In some embodiments, the amplification or the extension of the undesirable nucleic acid species is reduced by at least 50%. In some embodiments, the amplification or the extension of the undesirable nucleic acid species is reduced by at least 80%. In some embodiments, the amplification or the extension of the undesirable nucleic acid species is reduced by at least 90%. In some embodiments, the amplification or the extension of the undesirable nucleic acid species is reduced by at least 95%. In some embodiments, the amplification or the extension of the undesirable nucleic acid species is reduced by at least 99%.


In some embodiments, the blocking oligonucleotide is 10 nt to 50 nt long. In some embodiments, the blocking oligonucleotide is 20 nt to 30 nt long. In some embodiments, the blocking oligonucleotide is about 25 nt long.


In some embodiments, the one or more undesirable nucleic acid species amounts to about 50% of the nucleic acid content of the sample. In some embodiments, the one or more undesirable nucleic acid species amounts to about 60% of the nucleic acid content of the sample. In some embodiments, the one or more undesirable nucleic acid species amounts to about 70% of the nucleic acid content of the sample. In some embodiments, the one or more undesirable nucleic acid species amounts to about 80% of the nucleic acid content of the sample.


In some embodiments, the undesirable nucleic acid species is selected from the group consisting of rRNA, mtRNA, genomic DNA, intronic sequence, high abundance sequence, and any combination thereof. In some embodiments, the blocking oligonucleotides specifically bind to within 100 nt of the 3′ end of the one or more undesirable nucleic acid species. In some embodiments, the blocking oligonucleotides specifically bind to within 100 nt of the 5′ end of the one or more undesirable nucleic acid species. In some embodiments, the blocking oligonucleotides specifically bind to within 100 nt of the middle of the one or more undesirable nucleic acid species.


In some embodiments, the methods further comprise removing the hybridized complex formed between the blocking oligonucleotide and the undesirable nucleic acid species. In some embodiments, the removing comprises immobilizing the hybridized complex formed between the blocking oligonucleotide and the undesirable nucleic acid species on a solid support. In some embodiments, the blocking oligonucleotide comprises an affinity moiety. In some embodiments, solid support comprises a binding partner of the affinity moiety. In some embodiments, the affinity moiety is a functional group selected from the group consisting of biotin, streptavidin, heparin, an aptamer, a click-chemistry moiety, digoxigenin, primary amine(s), carboxyl(s), hydroxyl(s), aldehyde(s), ketone(s), and any combination thereof. In some embodiments, the affinity moiety is biotin. In some embodiments, the solid support comprises streptavidin.


In some embodiments, the amplifying comprises PCR amplification of the plurality of extension products. In some embodiments, each of the plurality of oligonucleotide probes comprises a cell label sequence, a sample label sequence, a location label sequence, a binding site for a universal primer, or any combination thereof. In some embodiments, the plurality of oligonucleotide probes comprises at least 100 different molecular label sequences. In some embodiments, the plurality of oligonucleotide probes comprises at least 1,000 different molecular label sequences. In some embodiments, the plurality of oligonucleotide probes comprises at least 10,000 different molecular label sequences. In some embodiments, the plurality of oligonucleotide probes comprises the same cell label sequence. In some embodiments, the plurality of amplicons comprises a cDNA library. In some embodiments, the sample comprises a single cell, a plurality of cells, a tissue sample, or any combination thereof. In some embodiments, the sample is a single cell.


In some embodiments, the methods further comprise sequencing the plurality of amplicons. In some embodiments, the undesirable nucleic acid species represents less than 50% of the plurality of amplicons. In some embodiments, the undesirable nucleic acid species represents less than 20% of the plurality of amplicons. In some embodiments, the undesirable nucleic acid species represents less than 10% of the plurality of amplicons. In some embodiments, the undesirable nucleic acid species represents less than 5% of the plurality of amplicons. In some embodiments, the plurality of oligonucleotide probes is immobilized on a substrate. In some embodiments, the substrate is a particle. In some embodiments, the substrate is a bead. In some embodiments, the plurality of nucleic acid target molecules comprises mRNA target molecules. In some embodiments, the binding region comprises poly-dT sequence.


Also disclosed herein is a kit for selective amplification of a plurality of nucleic acid target molecules in a sample. In some embodiments, the kit comprises: a plurality of oligonucleotide probes, wherein each of the plurality of oligonucleotide probes comprises a molecular label sequence and a binding region; and a plurality of blocking oligonucleotides that specifically binds to a plurality of undesirable nucleic acid species in the sample, wherein each blocking oligonucleotide probe is unable to function as a primer for a reverse transcriptase or a polymerase.


In some embodiments, the blocking oligonucleotide is a locked nucleic acid (LNA), a peptide nucleic acid (PNA), a DNA, an LNA/PNA chimera, an LNA/DNA chimera, or a PNA/DNA chimera. In some embodiments, the kit comprises blocking oligonucleotides that specifically binds to two or more undesirable nucleic acid species. In some embodiments, the kit comprises blocking oligonucleotides that specifically binds to at least 10 undesirable nucleic acid species. In some embodiments, the kit comprises blocking oligonucleotides that specifically binds to at least 100 undesirable nucleic acid species.


In some embodiments, the blocking oligonucleotide is 10 nt to 50 nt long. In some embodiments, the blocking oligonucleotide is 20 nt to 30 nt long. In some embodiments, the blocking oligonucleotide is about 25 nt long. In some embodiments, the undesirable nucleic acid species is selected from the group consisting of rRNA, mtRNA, genomic DNA, intronic sequence, high abundance sequence, and any combination thereof.


In some embodiments, the blocking oligonucleotides specifically bind to within 100 nt of the 3′ end of the undesirable nucleic acid species. In some embodiments, the blocking oligonucleotides specifically bind to within 100 nt of the 5′ end of the undesirable nucleic acid species. In some embodiments, the blocking oligonucleotides specifically bind to within 100 nt of the middle of the undesirable nucleic acid species. In some embodiments, the blocking oligonucleotide comprises an affinity moiety.


In some embodiments, each of the plurality of oligonucleotide probes comprises a cell label sequence, a sample label sequence, a location label sequence, a binding site for a universal primer, or any combination thereof. In some embodiments, the binding region comprises poly-dT. In some embodiments, the plurality of oligonucleotide probes is immobilized on a substrate. In some embodiments, the substrate is a particle, for example a bead.


In some embodiments, the kit further comprises an enzyme. The enzyme can be, for example, a reverse transcriptase, a polymerase, a ligase, a nuclease, and any combination thereof. In some embodiments, each blocking oligonucleotide probe has a Tm of at least 60° C. In some embodiments, the plurality of oligonucleotide probes comprises at least 100 different molecular label sequences. In some embodiments, the plurality of oligonucleotide probes comprises at least 1,000 different molecular label sequences. In some embodiments, the plurality of oligonucleotide probes comprises at least 10,000 different molecular label sequences. In some embodiments, the plurality of oligonucleotide probes comprises the same cell label sequence.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows a schematic illustration of an exemplary method for selective amplification using blocking oligonucleotides that specifically bind to undesirable nucleic acid species.





DETAILED DESCRIPTION
Definitions

Unless otherwise defined, all technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art in the field to which this disclosure belongs. As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Any reference to “or” herein is intended to encompass “and/or” unless otherwise stated.


As used herein the term “associated” or “associated with” can mean that two or more species are identifiable as being co-located at a point in time. An association can mean that two or more species are or were within a similar container. An association can be an informatics association, where for example digital information regarding two or more species is stored and can be used to determine that one or more of the species were co-located at a point in time. An association can also be a physical association. In some instances two or more associated species are “tethered”, “attached”, or “immobilized” to one another or to a common solid or semisolid surface. An association may refer to covalent or non-covalent means for attaching labels to solid or semi-solid supports such as beads. An association may comprise hybridization between a target and a label.


As used herein, the term “complementary” can refer to the capacity for precise pairing between two nucleotides. For example, if a nucleotide at a given position of a nucleic acid is capable of hydrogen bonding with a nucleotide of another nucleic acid, then the two nucleic acids are considered to be complementary to one another at that position. Complementarity between two single-stranded nucleic acid molecules may be “partial,” in which only some of the nucleotides bind, or it may be complete when total complementarity exists between the single-stranded molecules. A first nucleotide sequence can be said to be the “complement” of a second sequence if the first nucleotide sequence is complementary to the second nucleotide sequence. A first nucleotide sequence can be said to be the “reverse complement” of a second sequence, if the first nucleotide sequence is complementary to a sequence that is the reverse (i.e., the order of the nucleotides is reversed) of the second sequence. As used herein, the terms “complement”, “complementary”, and “reverse complement” can be used interchangeably. It is understood from the disclosure that if a molecule can hybridize to another molecule it may be the complement of the molecule that is hybridizing.


As used herein, the term “digital counting” can refer to a method for estimating a number of target molecules in a sample. Digital counting can include the step of determining a number of unique labels that have been associated with targets in a sample. This stochastic methodology transforms the problem of counting molecules from one of locating and identifying identical molecules to a series of yes/no digital questions regarding detection of a set of predefined labels.


As used herein, the term “label” or “labels” can refer to nucleic acid codes associated with a target within a sample. A label can be, for example, a nucleic acid label. A label can be an entirely or partially amplifiable label. A label can be entirely or partially sequencable label. A label can be a portion of a native nucleic acid that is identifiable as distinct. A label can be a known sequence. A label can comprise a junction of nucleic acid sequences, for example a junction of a native and non-native sequence. As used herein, the term “label” can be used interchangeably with the terms, “index”, “tag,” or “label-tag.” Labels can convey information. For example, in various embodiments, labels can be used to determine an identity of a sample, a source of a sample, an identity of a cell, and/or a target.


As used herein, a “nucleic acid” can generally refer to a polynucleotide sequence, or fragment thereof. A nucleic acid can comprise nucleotides. A nucleic acid can be exogenous or endogenous to a cell. A nucleic acid can exist in a cell-free environment. A nucleic acid can be a gene or fragment thereof. A nucleic acid can be DNA. A nucleic acid can be RNA. A nucleic acid can comprise one or more analogs (e.g. altered backgone, sugar, or nucleobase). Some non-limiting examples of analogs include: 5-bromouracil, peptide nucleic acid, xeno nucleic acid, morpholinos, locked nucleic acids, glycol nucleic acids, threose nucleic acids, dideoxynucleotides, cordycepin, 7-deaza-GTP, florophores (e.g. rhodamine or flurescein linked to the sugar), thiol containing nucleotides, biotin linked nucleotides, fluorescent base analogs, CpG islands, methyl-7-guanosine, methylated nucleotides, inosine, thiouridine, pseudourdine, dihydrouridine, queuosine, and wyosine. “Nucleic acid”, “polynucleotide, “target polynucleotide”, and “target nucleic acid” can be used interchangeably.


A nucleic acid can comprise one or more modifications (e.g., a base modification, a backbone modification), to provide the nucleic acid with a new or enhanced feature (e.g., improved stability). A nucleic acid can comprise a nucleic acid affinity tag. A nucleoside can be a base-sugar combination. The base portion of the nucleoside can be a heterocyclic base. The two most common classes of such heterocyclic bases are the purines and the pyrimidines. Nucleotides can be nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to the 2′, the 3′, or the 5′ hydroxyl moiety of the sugar. In forming nucleic acids, the phosphate groups can covalently link adjacent nucleosides to one another to form a linear polymeric compound. In turn, the respective ends of this linear polymeric compound can be further joined to form a circular compound; however, linear compounds are generally suitable. In addition, linear compounds may have internal nucleotide base complementarity and may therefore fold in a manner as to produce a fully or partially double-stranded compound. Within nucleic acids, the phosphate groups can commonly be referred to as forming the internucleoside backbone of the nucleic acid. The linkage or backbone of the nucleic acid can be a 3′ to 5′ phosphodiester linkage.


A nucleic acid can comprise a modified backbone and/or modified internucleoside linkages. Modified backbones can include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. Suitable modified nucleic acid backbones containing a phosphorus atom therein can include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates such as 3′-alkylene phosphonates, 5′-alkylene phosphonates, chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, phosphorodiamidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates, and boranophosphates having normal 3′-5′ linkages, 2′-5′ linked analogs, and those having inverted polarity wherein one or more internucleotide linkages is a 3′ to 3′, a 5′ to 5′ or a 2′ to 2′ linkage.


A nucleic acid can comprise polynucleotide backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These can include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; riboacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts.


A nucleic acid can comprise a nucleic acid mimetic. The term “mimetic” can be intended to include polynucleotides wherein only the furanose ring or both the furanose ring and the internucleotide linkage are replaced with non-furanose groups, replacement of only the furanose ring can also be referred as being a sugar surrogate. The heterocyclic base moiety or a modified heterocyclic base moiety can be maintained for hybridization with an appropriate target nucleic acid. One such nucleic acid can be a peptide nucleic acid (PNA). In a PNA, the sugar-backbone of a polynucleotide can be replaced with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleotides can be retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. The backbone in PNA compounds can comprise two or more linked aminoethylglycine units which gives PNA an amide containing backbone. The heterocyclic base moieties can be bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone.


A nucleic acid can comprise a morpholino backbone structure. For example, a nucleic acid can comprise a 6-membered morpholino ring in place of a ribose ring. In some of these embodiments, a phosphorodiamidate or other non-phosphodiester internucleoside linkage can replace a phosphodiester linkage.


A nucleic acid can comprise linked morpholino units (i.e. morpholino nucleic acid) having heterocyclic bases attached to the morpholino ring. Linking groups can link the morpholino monomeric units in a morpholino nucleic acid. Non-ionic morpholino-based oligomeric compounds can have less undesired interactions with cellular proteins. Morpholino-based polynucleotides can be nonionic mimics of nucleic acids. A variety of compounds within the morpholino class can be joined using different linking groups. A further class of polynucleotide mimetic can be referred to as cyclohexenyl nucleic acids (CeNA). The furanose ring normally present in a nucleic acid molecule can be replaced with a cyclohexenyl ring. CeNA DMT protected phosphoramidite monomers can be prepared and used for oligomeric compound synthesis using phosphoramidite chemistry. The incorporation of CeNA monomers into a nucleic acid chain can increase the stability of a DNA/RNA hybrid. CeNA oligoadenylates can form complexes with nucleic acid complements with similar stability to the native complexes. A further modification can include Locked Nucleic Acids (LNAs) in which the 2′-hydroxyl group is linked to the 4′ carbon atom of the sugar ring thereby forming a 2′-C,4′-C-oxymethylene linkage thereby forming a bicyclic sugar moiety. The linkage can be a methylene (—CH2-), group bridging the 2′ oxygen atom and the 4′ carbon atom wherein n is 1 or 2. LNA and LNA analogs can display very high duplex thermal stabilities with complementary nucleic acid (Tm=+3 to +10° C.), stability towards 3′-exonucleolytic degradation and good solubility properties.


A nucleic acid may also include nucleobase (often referred to simply as “base”) modifications or substitutions. As used herein, “unmodified” or “natural” nucleobases can include the purine bases, (e.g. adenine (A) and guanine (G)), and the pyrimidine bases, (e.g. thymine (T), cytosine (C) and uracil (U)). Modified nucleobases can include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl (—C═C—CH3) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 2-F-adenine, 2-aminoadenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine. Modified nucleobases can include tricyclic pyrimidines such as phenoxazine cytidine (1H-pyrimido(5,4-b)(1,4)benzoxazin-2(3H)-one), phenothiazine cytidine (1H-pyrimido(5,4-b)(1,4)benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g. 9-(2-aminoethoxy)-H-pyrimido(5,4-(b) (1,4)benzoxazin-2(3H)-one), carbazole cytidine (2H-pyrimido(4,5-b)indol-2-one), pyridoindole cytidine (Hpyrido(3′,′:4, 5)pyrrolo[2,3-d]pyrimidin-2-one).


As used herein, the term “sample” can refer to a composition comprising targets. Suitable samples for analysis by the disclosed methods, devices, and systems include cells, single cells, tissues, organs, or organisms.


As used herein, the term “sampling device” or “device” can refer to a device which may take a section of a sample and/or place the section on a substrate. A sample device can refer to, for example, an fluorescence activated cell sorting (FACS) machine, a cell sorter machine, a biopsy needle, a biopsy device, a tissue sectioning device, a microfluidic device, a blade grid, and/or a microtome.


As used herein, the term “solid support” can refer to discrete solid or semi-solid surfaces to which a plurality of stochastic barcodes may be attached. A solid support may encompass any type of solid, porous, or hollow sphere, ball, bearing, cylinder, or other similar configuration composed of plastic, ceramic, metal, or polymeric material (e.g., hydrogel) onto which a nucleic acid may be immobilized (e.g., covalently or non-covalently). A solid support may comprise a discrete particle that may be spherical (e.g., microspheres) or have a non-spherical or irregular shape, such as cubic, cuboid, pyramidal, cylindrical, conical, oblong, or disc-shaped, and the like. A plurality of solid supports spaced in an array may not comprise a substrate. A solid support may be used interchangeably with the term “bead.” As used herein, “solid support” and “substrate” can be used interchangeably.


As used herein, the term “stochastic barcode” refers to a polynucleotide sequence comprising labels of the present disclosure. A stochastic barcode can be a polynucleotide sequence that can be used for stochastic barcoding. Stochastic barcodes can be used to quantify targets within a sample. Stochastic barcodes can be used to control for errors which may occur after a label is associated with a target. For example, a stochastic barcode can be used to assess amplification or sequencing errors. A stochastic barcode associated with a target can be called a stochastic barcode-target or stochastic barcode-tag-target.


As used herein, the term “stochastic barcoding” refers to the random labeling (e.g., barcoding) of nucleic acids. Stochastic barcoding can utilize a recursive Poisson strategy to associate and quantify labels associated with targets. As used herein, the term “stochastic barcoding” can be used interchangeably with “stochastic labeling.”


As used here, the term “target” can refer to a composition which can be associated with a stochastic barcode. Exemplary suitable targets for analysis by the disclosed methods, devices, and systems include oligonucleotides, DNA, RNA, mRNA, microRNA, tRNA, and the like. Targets can be single or double stranded. In some embodiments targets can be proteins, polypeptides or peptides. In some embodiments targets are lipids. As used herein, “target” can be used interchangeably with “species”.


The term “reverse transcriptases” can refer to a group of enzymes having reverse transcriptase activity (i.e., that catalyze synthesis of DNA from an RNA template). In general, such enzymes include, but are not limited to, retroviral reverse transcriptase, retrotransposon reverse transcriptase, retroplasmid reverse transcriptases, retron reverse transcriptases, bacterial reverse transcriptases, group II intron-derived reverse transcriptase, and mutants, variants or derivatives thereof. Non-retroviral reverse transcriptases include non-LTR retrotransposon reverse transcriptases, retroplasmid reverse transcriptases, retron reverse transciptases, and group II intron reverse transcriptases. Examples of group II intron reverse transcriptases include the Lactococcus lactis Ll.LtrB intron reverse transcriptase, the Thermosynechococcus elongatus TeI4c intron reverse transcriptase, or the Geobacillus stearothermophilus GsI-IIC intron reverse transcriptase. Other classes of reverse transcriptases can include many classes of non-retroviral reverse transcriptases (i.e., retrons, group II introns, and diversity-generating retroelements among others).


Methods of Selective Amplification and/or Extension


Some embodiments disclosed herein provide methods of selective amplification and/or extension of a plurality of nucleic acid target molecules in a sample. The methods and compositions can, for example, reduce the amplification and/or extension of undesirable nucleic acid species in the sample, allow selective removal of undesirable nucleic acid species of the sample, or both.


For example, a sample can comprise a plurality of nucleic acid target molecules, and one or more undesirable nucleic acid species. In some embodiments, the method can significantly reduce the amplification, the extension, or both of the one or more undesirable nucleic acid species as compared to the plurality of nucleic acid target molecules in the sample. For example, the amplification and/or extension of the one or more undesirable nucleic acid species can be reduced by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99% or more in comparison to the amplification and/or extension of at least one of the nucleic acid target molecules in the sample, or the average amplification and/or extension of the one or more of the nucleic acid target molecules in the sample. In some embodiments, the methods disclosed herein can significantly reduce the amplification and/or extension of the one or more undesirable nucleic acid species as compared to the nucleic acid target molecules without significantly affecting the amplification and/or extension of the nucleic acid target molecules in the sample.


As used herein, a “nucleic acid species” refers to polynucleotides (for example, single-stranded polynucleotides) that are the same or substantially the same in sequence, or complement of one another, or are capable of hybridize to one another, or are transcripts from the same genetic locus, or encode the same protein or fragment thereof. In some embodiments, members of a nucleic acid species are at least 80%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% homologous to one another, or complement thereof. In some embodiments, members of a species can hybridize to one another under high stringent hybridization conditions. In some embodiments, members of a species can hybridize to one another under moderate stringent hybridization conditions. In some embodiments, members of a species can hybridize to one another under low stringent hybridization conditions. In some embodiments, members of a species are transcripts from the same genetic locus and the transcripts can be of the same or different length. The species is, in some embodiments, genomic DNA, ribosomal RNA (rRNA), mitochondrial DNA (mtDNA), cDNA, mRNA, or a combination thereof.


In some embodiments, the methods and compositions disclosed herein can reduce the amplification and/or extension of one or more undesirable nucleic acid species in a sample. For example, the methods and compositions disclosed herein can reduce the amplification and/or extension of at least 1, at least 2, at least 3, at least 4, at least 5, at least 10, at least 20, at least 50, at least 100, at least 200, at least 500, at least 1,000, or more, undesirable nucleic acid species in the sample. In some embodiments, the methods and compositions disclosed herein can reduce the amplification and/or extension by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99%, or 100% of each of the one or more undesirable nucleic acid species in the sample. In some embodiments, the methods and compositions disclosed herein abolish the amplification and/or extension of each of the one or more undesirable nucleic acid species in the sample. In some embodiments, the methods and compositions disclosed herein can reduce amplification and/or extension by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 99%, or 100% of at least one of the one or more undesirable nucleic acid species. In some embodiments, the methods and compositions disclosed herein abolish amplification and/or extension of at least one of the one or more undesirable nucleic acid species. In some embodiments, the methods and compositions disclosed herein reduce the amplification and/or extension of the total of undesirable nucleic acid species.


In some embodiments, the methods and compositions disclosed herein can reduce the amplification and/or extension of one or more undesirable nucleic acid species without significantly reducing amplification and/or extension of the nucleic acid target molecules in the same sample. For example, in some embodiments, the methods and compositions disclosed herein can reduce the amplification and/or extension by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% for each of the one or more undesirable nucleic acid species without significantly reducing amplification and/or extension of the nucleic acid target molecules. In some embodiments, the methods and compositions disclosed herein can reduce the amplification and/or extension by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the total of undesirable nucleic acid species without significantly reducing amplification and/or extension of the nucleic acid target molecules. In some embodiments, the methods and compositions disclosed herein can reduce the amplification and/or extension of one or more undesirable nucleic acid species while keeping at least at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the amplification and/or extension of each of the nucleic acid target molecules. In some embodiments, the methods and compositions disclosed herein can reduce the amplification and/or extension of one or more undesirable nucleic acid species while keeping at least at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the amplification and/or extension of at least one of the nucleic acid target molecules. In some embodiments, the methods and compositions disclosed herein can reduce the amplification and/or extension of one or more undesirable nucleic acid species while keeping at least at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the amplification and/or extension of the total of the nucleic acid target molecules.


As shown in FIG. 1, a sample comprises a whole transcriptome amplification (WTA) product 100 contains an undesirable nucleic acid species 105 and a nucleic acid target molecule 110. During an amplification reaction 120, an LNA/PNA blocking oligonucleotide 125 that specifically binds to the undesirable nucleic acid species 105 (binding shown at the 3′ end of 105 as a non-limiting example) is provided, which reduces or inhibits the amplification of the undesirable nucleic acid species 105. On the other hand, a copy 130 of the nucleic acid target molecule 110 is synthesized. In some embodiments, the amplification reaction 120 can be used in ending amplification step of WTA after end selection. In some embodiments, the hybridization complex between the undesirable nucleic acid species 105 and the blocking oligonucleotide 125 may be removed, for example, by immobilizing to a solid support. Completion 140 of the amplification reaction results in a library 150 comprising multiple copies 145 of the nucleic acid target molecule 110.


Nucleic Acid Target Molecules


In some embodiments, the methods disclosed herein comprise providing a sample comprising a plurality of nucleic acid target molecules. It would be appreciated by one of skill in the art that the plurality of nucleic acid target molecules can comprise a variety of nucleic acid target molecules. For example, the nucleic acid target molecules can comprise DNA molecules, RNA molecules, genomic DNA molecules, cDNA molecules, mRNA molecules, rRNA molecules, mtDNA, siRNA molecules, or any combination thereof. The nucleic acid target molecule can be double-stranded or single-stranded. In some embodiments, the plurality of nucleic acid target molecules can comprise polyA RNA molecules. In some embodiments, the plurality of nucleic acid target molecules comprise at least 100, at least 1,000, at least 10,000, at least 20,000, at least 30,000, at least 40,000, at least 50,000, at least 100,000, at least 1,000,000, or more nucleic acid species. In some embodiments, the plurality of nucleic acid target molecules can be from a sample, such as a single cell, a tissue, or a plurality of cells. In some embodiments, the plurality of nucleic acid target molecules can be pooled from a plurality of samples, such as a plurality of single cells or samples from different subjects (e.g., patients).


In some embodiments, the sample can comprise one or more undesirable nucleic acid species. As used herein, an “undesirable nucleic acid species” refers to a nucleic acid species that is present, e.g., in high amount, in a sample, for example the nucleic acid species representing 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 20%, 30%, 40%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, or more, or a range between any two of these values of the nucleic acid content in the sample. In some embodiments, the sample can comprise at least 1, at least 2, at least 3, at least 4, at least 5, at least 10, at least 20, at least 50, at least 100, at least 200, at least 500, at least 1,000, or more, undesirable nucleic acid species. In some embodiments, the total of all the undesirable nucleic acid species represent at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, or more of the nucleic acid content in the sample. In some embodiments, undesirable nucleic acid species can comprise polynucleotides encoding one or more ribosomal proteins. In some embodiments, undesirable nucleic acid species comprise rRNA. In some embodiments, undesirable nucleic acid species can comprise polynucleotides encoding one or more mitochondrial proteins. In some embodiments, undesirable nucleic acid species comprise mtDNA. In some embodiments, undesirable nucleic acid species can comprise polynucleotides encoding one or more housekeeping proteins. In some embodiments, undesirable nucleic acid species can comprise mRNA, rRNA, mtRNA, genomic DNA, intronic sequence, high abundance sequence, and any combination thereof.


In some embodiments, the plurality of nucleic acid target molecules comprises an unnormalized nucleic acid library, a partially normalized nucleic acid library, or a nucleic acid library that has been normalized by other methods, such as a cDNA library, a genomic DNA library, or the like. In some embodiments, the plurality of nucleic acid target molecules can comprise a pooled unnormalized nucleic acid library, such as a pooled unnormalized nucleic acid library constructed from a plurality of unnormalized nucleic acid libraries each representing a single cell. In some embodiments, the unnormalized nucleic acid library is a cDNA library. In some embodiments, the unnormalized nucleic acid library is a genomic library. In some embodiments, the unnormalized nucleic acid library is a single-cell nucleic acid library.


Blocking Oligonucleotides


In some embodiments, the methods disclosed herein comprise providing a blocking oligonucleotide that specifically binds to at least one of the one or more undesirable nucleic acid species. The blocking oligonucleotides can be provided at any point during the methods disclosed herein so that they can reduce the amplification and/or extension of the undesirable nucleic acid species. For example, the blocking oligonucleotides can be provided before, during or after the extension step, before or during the amplification step, before, during or after providing a plurality of oligonucleotide steps, before, during or after contacting the plurality of oligonucleotide probes with the plurality of nucleic acid target molecules for hybridization, or any combination thereof. A “blocking oligonucleotide” as used herein refers to a nucleic acid molecule that can specifically bind to at least one of the one or more undesirable nucleic acid species, whereby the specifically binding between the blocking oligonucleotide and the one or more undesirable nucleic acid species can reduce the amplification or extension (e.g., reverse transcription) of the one or more undesirable nucleic acid species. For example, the blocking oligonucleotide can comprise a nucleic acid sequence capable of hybridizing with one or more undesirable nucleic acid species. In some embodiments, a plurality of blocking oligonucleotides can be provided. The plurality of blocking oligonucleotides can specifically bind to at least 1, at least 2, at least 5, at least 10, at least 100, at least 1,000 or more of the one or more undesirable nucleic acid species. The location at which a blocking oligonucleotide specifically binds to an undesirable nucleic acid species can vary. For example, blocking oligonucleotide can specifically binds to a sequence close to the 5′ end of the undesirable nucleic acid species. In some embodiments, the blocking oligonucleotide can specifically bind to within 10 nt, 20 nt, 30 nt, 40 nt, 50 nt, 100 nt, 200 nt, 300 nt, 400 nt, 500 nt, or 1,000 nt of the 5′ end of at least one of the one or more undesirable nucleic acid species. In some embodiments, blocking oligonucleotide can specifically binds to a sequence close to the 3′ end of the undesirable nucleic acid species. For example, the blocking oligonucleotide can specifically bind to within 10 nt, 20 nt, 30 nt, 40 nt, 50 nt, 100 nt, 200 nt, 300 nt, 400 nt, 500 nt, 1,000 nt of the 3′ end of at least one of the one or more undesirable nucleic acid species. As another example, blocking oligonucleotide can specifically binds to a sequence in the middle portion of the undesirable nucleic acid species. In some embodiments, the blocking oligonucleotide can specifically bind to within 10 nt, 20 nt, 30 nt, 40 nt, 50 nt, 100 nt, 200 nt, 300 nt, 400 nt, 500 nt, 1,000 nt from the middle point of at least one of the one or more undesirable nucleic acid species.


In some embodiments, the specifically binding between the blocking oligonucleotide and the undesirable nucleic acid species can reduce the amplification and/or extension of the undesirable nucleic acid species by at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, at least 99%, or more.


It is contemplated that the blocking oligonucleotide may reduce the amplification and/or extension of the undesirable nucleic acid species by, for example, forming a hybridization complex with the undesirable nucleic acid species having a high melting temperature (Tm), by not being able to function as a primer for a reverse transcriptase or a polymerase, a combination thereof, etc. In some embodiments, the blocking oligonucleotide can have a Tm that is, is about, is at least, 50° C., 60° C., 65° C., 70° C., 75° C., 80° C., 85° C., 90° C., 95° C., or a range between any two of these values. In some embodiments, the blocking oligonucleotide can reduce the amplification and/or extension of the undesirable nucleic acid species by competing with the amplification and/or extension primers for hybridization with the undesirable nucleic acid species.


The blocking oligonucleotide can, in some embodiments, comprise one or more non-natural nucleotides. Non-natural nucleotides can be, for example, photolabile or triggerable nucleotides. Examples of non-natural nucleotides can include, but are not limited to, peptide nucleic acid (PNA), morpholino and locked nucleic acid (LNA), as well as glycol nucleic acid (GNA) and threose nucleic acid (TNA). In some embodiments, the blocking oligonucleotide is a chimeric oligonucleotide, such as an LNA/PNA/DNA chimera, an LNA/DNA chimera, a PNA/DNA chimera, a GNA/DNA chimera, a TNA/DNA chimera, or a combination thereof.


The melting temperature (Tm) of a blocking oligonucleotide can be modified, in some embodiments, by adjusting the length of the blocking oligonucleotide. For example, a blocking oligonucleotide can have a length that is, is about, is less than, is more than, 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 35 nt, 40 nt, 45 nt, 50 nt, 60 nt, 70 nt, 80 nt, 90 nt, 100 nt, 200 nt, or a range between any two of the above values.


In some embodiments, the Tm of a blocking oligonucleotide is modified by the number of DNA residues in the blocking oligonucleotide that comprises an LNA/DNA chimera or a PNA/DNA chimera. For example, a blocking oligonucleotide that comprises an LNA/DNA chimera or a PNA/DNA chimera can have a percentage of DNA residues that is, is about, is less than, is more than, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, or a range between any two of the above values.


In some embodiments, a blocking oligonucleotide can be designed to be incapable of functioning as a primer or probe for an amplification and/or extension reaction. For example, the blocking oligonucleotide may be incapable of function as a primer for a reverse transcriptase or a polymerase. For example, a blocking oligonucleotide that comprises an LNA/DNA chimera or a PNA/DNA chimera can be designed to have a certain percentage of LNA or PNA residues, or to have LNA or PNA residues on certain locations, such as close to or at the 3′ end, 5′ end, or in the middle portion of the oligonucleotide. In some embodiments, a blocking oligonucleotide that comprises an LNA/DNA chimera or a PNA/DNA chimera can have a percentage of LNA or PNA residues that is, is about, is less than, is more than, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, or a range between any two of the above values.


In some embodiments, the methods disclosed herein can comprise removing the hybridized complex formed between the blocking oligonucleotide and the undesirable nucleic acid species. For example, the blocking oligonucleotides can comprise an affinity moiety. The affinity moiety can be a functional group selected from the group consisting of biotin, streptavidin, heparin, an aptamer, a click-chemistry moiety, digoxigenin, primary amine(s), carboxyl(s), hydroxyl(s), aldehyde(s), ketone(s), and any combination thereof. In some embodiments, the affinity moiety is biotin. In some embodiments, the blocking oligonucleotide can be immobilized to a solid support having a binding partner for the affinity moiety through the affinity moiety. In some embodiments, the binding partner is streptavidin.


Oligonucleotide Probes


In some embodiments, the methods disclosed herein comprise providing a plurality of oligonucleotide probes, wherein each of the plurality of oligonucleotide probes comprises a molecular label sequence and a binding region. In some embodiments, the methods disclosed herein comprise contacting the plurality of oligonucleotide probes with the plurality of nucleic acid target molecules for hybridization. The oligonucleotide probes can comprise a binding region that hybridizes to one or more of the plurality of nucleic acid target molecules and one or more of the undesirable nucleic acid species. In some embodiments, the binding region can be target specific. For example, the binding region is configured to bind specific sequence(s). In some embodiments, the binding region can be target nonspecific. In some embodiments, the binding region comprises or consists of poly-dT sequence. In some embodiments, the oligonucleotide probes can comprise a stochastic barcode. In some embodiments, the oligonucleotide probes can comprise a molecular label sequence, a cell label sequence, a sample label sequence, a location label sequence, a binding site for a universal primer, or any combination thereof.


It is contemplated that the methods and compositions disclosed herein can be used in conjunction of molecular label sequences, for example, oligonucleotide probes that comprise molecular label sequences. Accordingly, in some embodiments, the species of nucleic acid molecules as disclosed herein can include polynucleotides in the plurality of nucleic acid molecules that are the same or the complement of one another, or are capable of hybridize to one another, or are transcripts from the same genetic locus, or encode the same protein or fragment thereof, etc., but that are associated with different molecular label sequences. It would be appreciated that molecular label sequences can be used to identify occurrences of a nucleic acid species.


A molecular label sequence can comprise a nucleic acid sequence that provides identifying information for the specific nucleic acid. A molecular label sequence can comprise a nucleic acid sequence that provides a counter for the specific occurrence of the target nucleic acid. A molecular label sequence can be, for example, 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, or more, or a range between any of these values, nucleotides in length. A molecular label sequence can be, for example, be at most about 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 15, 12, 10, 9, 8, 7, 6, 5, 4 or fewer nucleotides in length.


It would be appreciated that in some embodiments, the methods and compositions disclosed herein may reduce amplification of undesirable nucleic acid species without significantly reducing the number of different molecular label sequences associated with the other nucleic acid target molecules. For example, the methods and compositions disclosed herein can reduce amplification of undesirable nucleic acid species while retaining at least at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the different molecular label sequences associated with the other nucleic acid target molecules. In some embodiments, the methods and compositions disclosed herein can reduce amplification of undesirable nucleic acid species by at least at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or at least 99% while retaining at least at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the different molecular label sequences associated with the other nucleic acid target molecules. In some embodiments, reducing amplification of undesirable nucleic acid species does not significantly reduce the number of different molecular label sequences associated with the other nucleic acid target molecules.


Extension


One or more extension reactions can be performed using the oligonucleotide probes that are hybridized to the plurality of nucleic acid target molecules to generate a plurality of extension products. In some embodiments, the oligonucleotide probes function as primers for the extension reaction, such as reverse transcription. The extension reactions can be performed with or without the presence of blocking oligonucleotides. In embodiments where blocking oligonucleotides are present in the extension reactions, the blocking oligonucleotides can reduce the extension of one or more undesirable nucleic acid species to which the blocking oligonucleotides specifically bind. In some embodiments, the blocking oligonucleotides do not significantly reduce the extension of the nucleic acid target molecules.


The plurality of nucleic acid target molecules can, in some embodiments, randomly associate with the oligonucleotide probes. Association can, for example, comprise hybridization of an oligonucleotide probe's binding region to a complementary portion of the target nucleic acid molecule (e.g., oligo dT sequence of the stochastic barcode can interact with a poly-A tail of a target nucleic acid molecule). The assay conditions used for hybridization (e.g. buffer pH, ionic strength, temperature, etc.) can be chosen to promote formation of specific, stable hybrids.


The disclosure provides for methods of associating a molecular label with a target nucleic acid using reverse transcription.


Amplification


In some embodiments, the methods disclosed herein can comprise amplifying a sample wherein the sample comprises a plurality of nucleic acid target molecules and one or more undesirable nucleic acid species, or amplifying the plurality of extension products to generate a plurality of amplicons. In some embodiments, one or more nucleic acid amplification reactions can be performed to create multiple copies of the target nucleic acid molecules or the extension products. In some embodiments, primers can be added for the amplification reaction, such as PCR. The amplification reactions can be performed in or without the presence of blocking oligonucleotides. In embodiments, where blocking oligonucleotides are present in the amplification reactions, the blocking oligonucleotides can reduce the amplification of one or more undesirable nucleic acid species to which the blocking oligonucleotides specifically bind. In some embodiments, the blocking oligonucleotides do not significantly reduce the amplification of the nucleic acid target molecules.


Amplification can be performed, in some embodiments, in a multiplexed manner, wherein multiple target nucleic acid sequences are amplified simultaneously. The amplification reaction can be used, for example, to add sequencing adaptors to the nucleic acid molecules. The amplification reactions can comprise amplifying at least a portion of a sample label, if present. The amplification reactions can comprise amplifying at least a portion of the cellular and/or molecular label. The amplification reactions can comprise amplifying at least a portion of a sample tag, a cell label, a spatial label, a molecular label, a target nucleic acid, or a combination thereof. The amplification reactions can, for example, comprise amplifying at least 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, or 100% of the plurality of target nucleic acids. The method may further comprise conducting one or more cDNA synthesis reactions to produce one or more cDNA copies of target-barcode molecules comprising a sample label, a cell label, a spatial label, and/or a molecular label.


In some embodiments, amplification can be performed using a polymerase chain reaction (PCR). As used herein, “PCR” refers to a reaction for the in vitro amplification of specific DNA sequences by the simultaneous primer extension of complementary strands of DNA. As used herein, PCR encompass derivative forms of the reaction, including but not limited to, RT-PCR, real-time PCR, nested PCR, quantitative PCR, multiplexed PCR, digital PCR, and assembly PCR.


Amplification of the labeled nucleic acids can comprise non-PCR based methods. Examples of non-PCR based methods include, but are not limited to, multiple displacement amplification (MDA), transcription-mediated amplification (TMA), whole transcriptome amplification (WTA), whole genome amplification (WGA), nucleic acid sequence-based amplification (NASBA), strand displacement amplification (SDA), real-time SDA, rolling circle amplification, or circle-to-circle amplification. Other non-PCR-based amplification methods include multiple cycles of DNA-dependent RNA polymerase-driven RNA transcription amplification or RNA-directed DNA synthesis and transcription to amplify DNA or RNA targets, a ligase chain reaction (LCR), and a Qβ replicase (Qβ) method, use of palindromic probes, strand displacement amplification, oligonucleotide-driven amplification using a restriction endonuclease, an amplification method in which a primer is hybridized to a nucleic acid sequence and the resulting duplex is cleaved prior to the extension reaction and amplification, strand displacement amplification using a nucleic acid polymerase lacking 5′ exonuclease activity, rolling circle amplification, and ramification extension amplification (RAM). In some instances, the amplification may not produce circularized transcripts.


In some instances, the methods disclosed herein further comprise conducting a polymerase chain reaction on the labeled nucleic acid (e.g., labeled-RNA, labeled-DNA, labeled-cDNA) to produce a labeled amplicon. The labeled amplicon can, for example, be a double-stranded molecule. The double-stranded molecule can comprise a double-stranded RNA molecule, a double-stranded DNA molecule, or a RNA molecule hybridized to a DNA molecule. One or both of the strands of the double-stranded molecule may comprise a sample label, a spatial label, a cell label, and/or a molecular label. The labeled amplicon can be a single-stranded molecule. The single-stranded molecule can comprise DNA, RNA, or a combination thereof. The nucleic acids of the disclosure comprise synthetic or altered nucleic acids.


Amplification can, for example, comprise use of one or more non-natural nucleotides. Non-natural nucleotides may comprise photolabile or triggerable nucleotides. Examples of non-natural nucleotides can include, but are not limited to, peptide nucleic acid (PNA), morpholino and locked nucleic acid (LNA), as well as glycol nucleic acid (GNA) and threose nucleic acid (TNA). Non-natural nucleotides may be added to one or more cycles of an amplification reaction. The addition of the non-natural nucleotides can be, for example, used to identify products as specific cycles or time points in the amplification reaction.


As described herein, conducting the one or more amplification reactions can comprise the use of one or more primers. A primer can, for example, comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more nucleotides. In some embodiments, the primer comprises at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 or more nucleotides. For example, the primer can comprise 12 to 15 nucleotides. The one or more primers can, for example, anneal to at least a portion of the plurality of labeled target nucleic acid molecules and oligonucleotides. For example, the one or more primers can anneal to the 3′ end or 5′ end of the plurality of labeled target nucleic acid molecules and oligonucleotides. The one or more primers can, in some embodiments, anneal to an internal region of the plurality of labeled target nucleic acid molecules and oligonucleotides. The internal region of a oligonucleotide or target nucleic acid molecule can be, for example, at least about 50, 100, 150, 200, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350, 360, 370, 380, 390, 400, 410, 420, 430, 440, 450, 460, 470, 480, 490, 500, 510, 520, 530, 540, 550, 560, 570, 580, 590, 600, 650, 700, 750, 800, 850, 900, or 1000 nucleotides from the 3′ ends and/or 5′ end of the oligonucleotide or the target nucleic acid molecule. The one or more primers may comprise a fixed panel of primers. The one or more primers may comprise at least one or more custom primers. The one or more primers may comprise at least one or more control primers. The one or more primers may comprise at least one or more gene-specific primers.


The one or more primers can comprise any universal primer of the disclosure. The universal primer may anneal to a universal primer binding site. The one or more custom primers can, in some embodiments, anneal to a first sample label, a second sample label, a spatial label, a cell label, a molecular label, a target, or any combination thereof. The one or more primers may comprise a universal primer and a custom primer.


Any amplification scheme can be used in the methods of the present disclosure. For example, in one scheme, the first round PCR can amplify molecules (e.g., attached to the bead) using a gene specific primer and a primer against the universal Illumina sequencing primer 1 sequence. The second round of PCR can amplify the first PCR products using a nested gene specific primer flanked by Illumina sequencing primer 2 sequence, and a primer against the universal Illumina sequencing primer 1 sequence. The third round of PCR adds P5 and P7 and sample index to turn PCR products into an Illumina sequencing library. Sequencing using 150 bp×2 sequencing can reveal the cell label and molecular index on read 1, the gene on read 2, and the sample index on index 1 read.


Amplification can be performed in one or more rounds. In some instances there are multiple rounds of amplification. Amplification can comprise two or more rounds of amplification. The first amplification can be an extension off X′ to generate the gene specific region. The second amplification can occur when a sample nucleic hybridizes to the newly generated strand.


In some embodiments, hybridization does not need to occur at the end of a nucleic acid molecule. In some embodiments, a target nucleic acid within an intact strand of a longer nucleic acid is hybridized and amplified. For example a target within a longer section of genomic DNA or mRNA. A target can be more than 50 nt, more than 100 nt, or more that 1000 nt from one end (e.g., 5′ end or 3′ end) of a polynucleotide.


Sequencing


In some embodiments, the extension products and/or the amplification products disclosed herein may be used for sequencing. Any suitable sequencing method known in the art can be used, preferably high-throughput approaches. For example, cyclic array sequencing using platforms such as Roche 454, Illumina Solexa, ABI-SOLiD, ION Torrent, Complete Genomics, Pacific Bioscience, Helicos, or the Polonator platform, may also be utilized. Sequencing may comprise MiSeq sequencing and/or HiSeq sequencing. The selective extension and/or amplification methods disclosed herein can, in some embodiments, increase the efficiency of sequencing by decreasing the number of sequencing reads for the undesirable nucleic acid species.


In some embodiments, after using the selective extension and/or amplification methods described herein, the sequencing reads for the undesirable nucleic acid species are less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5%, or less, of the total sequencing reads. In some embodiments, the sequencing reads for the undesirable nucleic acid species are less than 40% of the total sequencing reads. In some embodiments, the sequencing reads for the undesirable nucleic acid species are less than 30% of the total sequencing reads. In some embodiments, the sequencing reads for the undesirable nucleic acid species are less than 20% of the total sequencing reads. In some embodiments, the sequencing reads for the undesirable nucleic acid species are less than 10% of the total sequencing reads. In some embodiments, after using the selective extension and/or amplification methods described herein, the sequencing reads for the undesirable nucleic acid species are reduced to less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5% of the sequencing reads for the undesirable nucleic acid without using the selective extension and/or amplification methods described herein. In some embodiments, after using the selective extension and/or amplification methods described herein, the sequencing reads for the undesirable nucleic acid species are reduced to, or to about, 60%, 50%, 40%, 30%, 20%, 10%, 5%, 2%, 1%, 0.5%, or a range between any two of these values, of the sequencing reads for the undesirable nucleic acid without using the selective extension and/or amplification methods described herein.


In some embodiments, the methods and compositions disclosed herein can improve sequencing efficiency by decreasing the sequencing reads:molecular label ratio of an undesirable nucleic acid species and/or increasing the sequencing reads:molecular label ratio of a nucleic acid target molecule. For example, the ratio of sequencing reads to molecular label for an undesirable nucleic acid species can be less than 20, less than 15, less than 10, less than 9, less than 8, less than 7, less than 6, less than 5, less than 4, less than 3, less than 2, or less than 1. In some embodiments, the ratio of sequencing reads to molecular label for an undesirable nucleic acid species is 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, or a range between any two of these values.


Kits


Disclosed herein are kits for selective amplification and/or extension of a plurality of nucleic acid target molecules in a sample, wherein the sample comprises a plurality of target nucleic acid species and one or more undesirable nucleic acid species. In some embodiments, the kit comprises a plurality of oligonucleotide probes, wherein each of the plurality of oligonucleotide probes comprises a molecular label sequence and a binding region; and a plurality of blocking oligonucleotides that specifically binds to a plurality of undesirable nucleic acid species in the sample, wherein each blocking oligonucleotide probe is unable to function as a primer for a reverse transcriptase or a polymerase.


In some embodiments, the kit further comprises a plurality of blocking oligonucleotides. The plurality of blocking oligonucleotides can, for example, specifically bind to at least 1, at least 2, at least 5, at least 10, at least 100, at least 1,000 or more undesirable nucleic acid species in the sample. In some embodiments, the blocking oligonucleotide can specifically bind to within 10 nt, 20 nt, 30 nt, 40 nt, 50 nt, 100 nt, 200 nt, 300 nt, 400 nt, 500 nt, 1,000 nt of the 5′ end of the one or more undesirable nucleic acid species. In some embodiments, the blocking oligonucleotide can specifically bind to within 10 nt, 20 nt, 30 nt, 40 nt, 50 nt, 100 nt, 200 nt, 300 nt, 400 nt, 500 nt, 1,000 nt of the 3′ end of the one or more undesirable nucleic acid species. In some embodiments, the blocking oligonucleotide can specifically bind to within 10 nt, 20 nt, 30 nt, 40 nt, 50 nt, 100 nt, 200 nt, 300 nt, 400 nt, 500 nt, 1,000 nt surrounding the middle point of the one or more undesirable nucleic acid species.


It is contemplated that the blocking oligonucleotide may reduce the amplification and/or extension of the undesirable nucleic acid species by forming a hybridization complex with the undesirable nucleic acid species having a high melting temperature (Tm), by not being able to function as a primer for a reverse transcriptase or a polymerase, a combination thereof, etc. In some embodiments, the blocking oligonucleotide can have a Tm that is, is about, is at least, 50° C., 60° C., 65° C., 70° C., 75° C., 80° C., 85° C., 90° C., 95° C., or a range between any two of the above values. In some embodiments, the blocking oligonucleotide may reduce the amplification and/or extension of the undesirable nucleic acid species by competing with the amplification and/or extension primers for hybridization with the undesirable nucleic acid species.


The blocking oligonucleotide can, in some embodiments, comprise one or more non-natural nucleotides. Non-natural nucleotides can comprise photolabile or triggerable nucleotides. Examples of non-natural nucleotides can include, but are not limited to, peptide nucleic acid (PNA), morpholino and locked nucleic acid (LNA), as well as glycol nucleic acid (GNA) and threose nucleic acid (TNA). In some embodiments, the blocking oligonucleotide is a chimeric oligonucleotide, such as an LNA/PNA/DNA chimera, an LNA/DNA chimera, a PNA/DNA chimera, a GNA/DNA chimera, a TNA/DNA chimera, and a combination thereof.


It would be appreciated that the Tm of a blocking oligonucleotide can be modified by adjusting the length of the blocking oligonucleotide. For example, a blocking oligonucleotide can have a length that is, is about, is less than, is more than, 10 nt, 15 nt, 20 nt, 25 nt, 30 nt, 35 nt, 40 nt, 45 nt, 50 nt, 60 nt, 70 nt, 80 nt, 90 nt, 100 nt, 200 nt, or a range between any two of the above values.


In some embodiments, the Tm of a blocking oligonucleotide can be modified by adjusting the number of DNA residues in the blocking oligonucleotide that comprises an LNA/DNA chimera or a PNA/DNA chimera. For example, a blocking oligonucleotide that comprises an LNA/DNA chimera or a PNA/DNA chimera can have a percentage of DNA residues that is, is about, is less than, is more than, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, or a range between any two of the above values.


In some embodiments, a blocking oligonucleotide can be designed to being incapable of functioning as a primer for an extension or amplication. For example, the blocking oligonucleotide may be incapable of functioning as a primer for a reverse transcriptase, a polymerase, or both. For example, a blocking oligonucleotide that comprises an LNA/DNA chimera or a PNA/DNA chimera can be designed to have a certain percentage of LNA or PNA residues, or to have LNA or PNA residues at certain location(s), such as the 3′ end, the 5′ end, the internal region, or a combination thereof of the blocking oligonucleotide. In some embodiments, a blocking oligonucleotide that comprises an LNA/DNA chimera or a PNA/DNA chimera can have a percentage of LNA or PNA residues that is, is about, is less than, is more than, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, or a range between any two of the above values.


In some embodiments, the blocking oligonucleotides can comprise an affinity moiety. The affinity moiety can be a functional group selected from the group consisting of biotin, streptavidin, heparin, an aptamer, a click-chemistry moiety, digoxigenin, primary amine(s), carboxyl(s), hydroxyl(s), aldehyde(s), ketone(s), and any combination thereof. In some embodiments, the blocking oligonucleotides can be immobilized to a solid support having a binding partner for the affinity moiety through the affinity moiety.


In some embodiments, each of the oligonucleotide probes can comprise a molecular label, a cell label, a sample label, or any combination thereof. In some embodiments, each of the oligonucleotides can comprise a linker. In some embodiments, each of the oligonucleotide probes can comprise a binding site for an oligonucleotide probe, such as a poly A tail. For example, the poly A tail can be, e.g., oligodA18 (unanchored to a solid support) or oligoA18V (anchored to a solid support). The oligonucleotide probes can comprise DNA residues, RNA residues, or both.


In some embodiments, the kits can further comprise an enzyme. In some embodiments, the enzyme can be a reverse transcriptase, a polymerase, a ligase, a nuclease, or, any combination thereof.


Stochastic Barcodes


The oligonucleotide probes disclosed herein can comprise, or consists of, stochastic barcodes. As disclosed herein, a stochastic barcode can be a polynucleotide sequence that may be used to stochastically label (e.g., barcode, tag) a target. A stochastic barcode can comprise one or more labels. Exemplary labels include, but are not limited to, universal labels, cell labels, molecular labels, sample labels, plate labels, spatial labels, pre-spatial labels, and any combination thereof. A stochastic barcode can comprise a 5′ amine that may link the stochastic barcode to a solid support. The stochastic barcode can comprise one or more of a universal label, a dimension label, a spatial label, a cell label, and a molecular label. The universal label can be 5′-most label. The molecular label can be the 3′-most label. The spatial label, dimension label, and the cell label can be in any order. In some instances, the universal label, the spatial label, the dimension label, the cell label, and the molecular label are in any order. The stochastic barcode can comprise a target-binding region. The target-binding region can interact with a target (e.g., target nucleic acid, RNA, mRNA, DNA) in a sample. For example, a target-binding region can comprise an oligo dT sequence which can interact with poly-A tails of mRNAs. In some instances, the labels of the stochastic barcode (e.g., universal label, dimension label, spatial label, cell label, and molecular label) may be separated by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, a range between any two of these values, or more nucleotides.


A stochastic barcode can comprise one or more universal labels. The one or more universal labels can be the same for all stochastic barcodes in the set of stochastic barcodes (e.g., attached to a given solid support). In some embodiments, the one or more universal labels can be the same for all stochastic barcodes attached to a plurality of beads. In some embodiments, a universal label comprises a nucleic acid sequence that is capable of hybridizing to a sequencing primer. Sequencing primers can be used for sequencing stochastic barcodes comprising a universal label. Sequencing primers (e.g., universal sequencing primers) can comprise sequencing primers associated with high-throughput sequencing platforms. In some embodiments, a universal label may comprise a nucleic acid sequence that is capable of hybridizing to a PCR primer. In some embodiments, the universal label comprises a nucleic acid sequence that is capable of hybridizing to a sequencing primer and a PCR primer. The nucleic acid sequence of the universal label that is capable of hybridizing to a sequencing or PCR primer may be referred to as a primer binding site. A universal label can comprise a sequence that may be used to initiate transcription of the stochastic barcode. A universal label can comprise a sequence that may be used for extension of the stochastic barcode or a region within the stochastic barcode. A universal label can be, or be at least about, 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length. A universal label can comprise at least about 10 nucleotides. A universal label can be at most about 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length. In some embodiments, a cleavable linker or modified nucleotide is part of the universal label sequence to enable the stochastic barcode to be cleaved off from the support. As used herein, a universal label can be used interchangeably with “universal PCR primer.”


A stochastic barcode can comprise a dimension label. A dimension label can comprise a nucleic acid sequence that provides information about a dimension in which the stochastic labeling occurred. For example, a dimension label can provide information about the time at which a target was stochastically barcoded. A dimension label can be associated with a time of stochastic barcoding in a sample. A dimension label can activated at the time of stochastic labeling. Different dimension labels can be activated at different times. The dimension label provides information about the order in which targets, groups of targets, and/or samples were stochastically barcoded. For example, a population of cells can be stochastically barcoded at the G0 phase of the cell cycle. The cells can be pulsed again with stochastic barcodes at the G1 phase of the cell cycle. The cells can be pulsed again with stochastic barcodes at the S phase of the cell cycle, and so on. Stochastic barcodes at each pulse (e.g., each phase of the cell cycle), can comprise different dimension labels. In this way, the dimension label provides information about which targets were labelled at which phase of the cell cycle. Dimension labels can interrogate many different biological times. Exemplary biological times can include, but are not limited to, the cell cycle, transcription (e.g., transcription initiation), and transcript degradation. In another example, a sample (e.g., a cell, a population of cells) can be stochastically labeled before and/or after treatment with a drug and/or therapy. The changes in the number of copies of distinct targets can be indicative of the sample's response to the drug and/or therapy.


In some embodiments, a dimension label is activatable. An activatable dimension label can be activated, for example, at a specific timepoint. The activatable dimension label can be constitutively activated (e.g., not turned off). The activatable dimension label can be reversibly activated (e.g., the activatable dimension label can be turned on and turned off). The dimension label can be reversibly activatable at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more times. The dimension label can be reversibly activatable 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 or more times. For example, the dimension label can be activated with fluorescence, light, a chemical event (e.g., cleavage, ligation of another molecule, addition of modifications (e.g., pegylated, sumoylated, acetylated, methylated, deacetylated, demethylated), a photochemical event (e.g., photocaging), and introduction of a non-natural nucleotide.


The dimension label can be identical for all stochastic barcodes attached to a given solid support (e.g., bead), but different for different solid supports (e.g., beads). In some embodiments, at least 60%, 70%, 80%, 85%, 90%, 95%, 97%, 99% or 100% of stochastic barcodes on the same solid support comprise the same dimension label. In some embodiments, at least 60% of stochastic barcodes on the same solid support comprise the same dimension label. In some embodiments, at least 95% of stochastic barcodes on the same solid support comprise the same dimension label.


There can be as many as 106 or more unique dimension label sequences represented in a plurality of solid supports (e.g., beads). A dimension label can, for example, be or be at least about 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length. A dimension label can be at most about 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 15, 12, 10, 9, 8, 7, 6, 5, 4 or fewer or more nucleotides in length. A dimension label can, for example, is about 5 to about 200 nucleotides, or about 10 to about 150 nucleotides in length. In some embodiments, a dimension label is from about 20 to about 125 nucleotides in length.


A stochastic barcode can comprise a spatial label. A spatial label can comprise a nucleic acid sequence that provides information about the spatial orientation of a target molecule which is associated with the stochastic barcode. A spatial label can be associated with a coordinate in a sample. The coordinate can be a fixed coordinate. For example a coordinate can be fixed in reference to a substrate. A spatial label can be in reference to a two or three-dimensional grid. A coordinate can be fixed in reference to a landmark. The landmark can be identifiable in space. A landmark can be a structure which can be imaged. A landmark can be a biological structure, for example an anatomical landmark. A landmark can be a cellular landmark, for instance an organelle. A landmark can be a non-natural landmark such as a structure with an identifiable identifier such as a color code, bar code, magnetic property, fluorescents, radioactivity, or a unique size or shape. A spatial label can be associated with a physical partition (e.g. a well, a container, or a droplet). In some instances, multiple spatial labels are used together to encode one or more positions in space.


The spatial label can be identical for all stochastic barcodes attached to a given solid support (e.g., bead), but different for different solid supports (e.g., beads). In some embodiments, at least 60%, 70%, 80%, 85%, 90%, 95%, 97%, 99% or 100% of stochastic barcodes on the same solid support comprise the same spatial label. In some embodiments, at least 60% of stochastic barcodes on the same solid support comprise the same spatial label. In some embodiments, at least 95% of stochastic barcodes on the same solid support comprise the same spatial label.


There can be as many as 106 or more unique spatial label sequences represented in a plurality of solid supports (e.g., beads). A spatial label can be, or be at least about, 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length. In some embodiments, a spatial label is most about 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 15, 12, 10, 9, 8, 7, 6, 5, 4 nucleotides in length. A spatial label can be, for example, from about 5 to about 200 nucleotides in length. A spatial label can be, for example, from about 10 to about 150 nucleotides in length. A spatial label can be from about 20 to about 125 nucleotides in length.


Stochastic barcodes can comprise a cell label. A cell label can comprise a nucleic acid sequence that provides information for determining which target nucleic acid originated from which cell. In some embodiments, the cell label is identical for all stochastic barcodes attached to a given solid support (e.g., bead), but different for different solid supports (e.g., beads). In some embodiments, at least 60%, 70%, 80%, 85%, 90%, 95%, 97%, 99% or 100% of stochastic barcodes on the same solid support comprise the same cell label. In some embodiments, at least 60% of stochastic barcodes on the same solid support comprise the same cell label. In some embodiment, at least 95% of stochastic barcodes on the same solid support comprise the same cell label.


There can be as many as 106 or more unique cell label sequences represented in a plurality of solid supports (e.g., beads). A cell label may be at least about 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length. A cell label can be, or be at most about, 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 15, 12, 10, 9, 8, 7, 6, 5, 4 or fewer or more nucleotides in length. A cell label can be, for example, from about 5 to about 200 nucleotides in length. A cell label can be, for example, from about 10 to about 150 nucleotides in length. A cell label can be, for example, from about 20 to about 125 nucleotides in length.


Stochastic barcodes can comprise a molecular label. A molecular label can comprise a nucleic acid sequence that provides identifying information for the specific type of target nucleic acid species hybridized to the stochastic barcode. A molecular label can comprise a nucleic acid sequence that provides a counter for the specific occurrence of the target nucleic acid species hybridized to the stochastic barcode (e.g., target-binding region). In some embodiments, a diverse set of molecular labels are attached to a given solid support (e.g., bead). In some embodiments, there can be as many as 106 or more unique molecular label sequences attached to a given solid support (e.g., bead). In some embodiments, there can be as many as 105 or more unique molecular label sequences attached to a given solid support (e.g., bead). In some embodiments, there can be as many as 104 or more unique molecular label sequences attached to a given solid support (e.g., bead). In some embodiments, there can be as many as 103 or more unique molecular label sequences attached to a given solid support (e.g., bead). In some embodiments, there can be as many as 102 or more unique molecular label sequences attached to a given solid support (e.g., bead). A molecular label can be at least about 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length. A molecular label can be at most about 300, 200, 100, 90, 80, 70, 60, 50, 40, 30, 20, 15, 12, 10, 9, 8, 7, 6, 5, 4 or fewer nucleotides in length.


Stochastic barcodes can comprise a target binding region. In some embodiments, the target binding regions comprise a nucleic acid sequence that hybridizes specifically to a target (e.g., target nucleic acid, target molecule, e.g., a cellular nucleic acid to be analyzed), for example to a specific gene sequence. In some embodiments, a target binding region comprise a nucleic acid sequence that may attach (e.g., hybridize) to a specific location of a specific target nucleic acid. In some embodiments, the target binding region comprise a nucleic acid sequence that is capable of specific hybridization to a restriction site overhang (e.g. an EcoRI sticky-end overhang). The stochastic barcode may then ligate to any nucleic acid molecule comprising a sequence complementary to the restriction site overhang.


A stochastic barcode can comprise a target-binding region. A target-binding region can hybridize with a target of interest. For example, a target-binding region can comprise an oligo dT which can hybridize with mRNAs comprising poly-adenylated ends. A target-binding region can be gene-specific. For example, a target-binding region can be configured to hybridize to a specific region of a target. A target-binding region can be, or be at least, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26 27, 28, 29, or 30 or more nucleotides in length. A target-binding region can be at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26 27, 28, 29, or 30 or more nucleotides in length. A target-binding region can be from 5-30 nucleotides in length. When a stochastic barcode comprises a gene-specific target-binding region, the stochastic barcode can be referred to as a gene-specific stochastic barcode.


A target binding region can comprise a non-specific target nucleic acid sequence. A non-specific target nucleic acid sequence can refer to a sequence that may bind to multiple target nucleic acids, independent of the specific sequence of the target nucleic acid. For example, target binding region can comprise a random multimer sequence, or an oligo-dT sequence that hybridizes to the poly-A tail on mRNA molecules. A random multimer sequence can be, for example, a random dimer, trimer, quatramer, pentamer, hexamer, septamer, octamer, nonamer, decamer, or higher multimer sequence of any length. In some embodiments, the target binding region is the same for all stochastic barcodes attached to a given bead. In some embodiments, the target binding regions for the plurality of stochastic barcodes attached to a given bead comprise two or more different target binding sequences. A target binding region can be, or be at least about, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length. In some embodiments, a target binding region is at most about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length.


A stochastic barcode can comprise an orientation property which can be used to orient (e.g., align) the stochastic barcodes. A stochastic barcode can comprise a moiety for isoelectric focusing. Different stochastic barcodes can comprise different isoelectric focusing points. When these stochastic barcodes are introduced to a sample, the sample can undergo isoelectric focusing in order to orient the stochastic barcodes into a known way. In this way, the orientation property can be used to develop a known map of stochastic barcodes in a sample. Exemplary orientation properties include, but are not limited to, electrophoretic mobility (e.g., based on size of the stochastic barcode), isoelectric point, spin, conductivity, and/or self-assembly. For example, stochastic barcodes can comprise an orientation property of self-assembly, can self-assemble into a specific orientation (e.g., nucleic acid nanostructure) upon activation.


The cell label and/or any label of the disclosure can further comprise a unique set of nucleic acid sub-sequences of defined length, e.g. 7 nucleotides each (equivalent to the number of bits used in some Hamming error correction codes), which are designed to provide error correction capability. Hamming codes, like other error-correcting codes, are based on the principle of redundancy and can be constructed by adding redundant parity bits to data that is to be transmitted over a noisy medium. Such error-correcting codes can encode sample identifiers with redundant parity bits, and “transmit” these sample identifiers as codewords. A Hamming code can refer an arithmetic process that identifies unique binary codes based upon inherent redundancy that are capable of correcting single bit errors. For example, a Hamming code can be matched with a nucleic acid barcode in order to screen for single nucleotide errors occurring during nucleic acid amplification. The identification of a single nucleotide error by using a Hamming code, thereby can allow for the correction of the nucleic acid barcode.


Hamming codes can be represented by a subset of the possible codewords that are chosen from the center of multidimensional spheres (i.e., for example, hyperspheres) in a binary subspace. Single bit errors may fall within hyperspheres associated with a specific codeword and can thus be corrected. On the other hand, double bit errors that do not associate with a specific codeword can be detected, but not corrected. Consider a first hypersphere centered at coordinates (0, 0, 0) (i.e., for example, using an x-y-z coordinate system), wherein any single-bit error can be corrected by falling within a radius of 1 from the center coordinates; i.e., for example, single bit errors having the coordinates of (0, 0, 0); (0, 1, 0); (0, 0, 1); (1, 0, 0), or (1, 1, 0). Likewise, a second hypersphere may be constructed wherein single-bit errors can be corrected by falling within a radius of 1 of its center coordinates (1, 1, 1) (i.e., for example, (1, 1, 1); (1, 0, 1); (0, 1, 0); or (0, 1, 1).


In some embodiments, the length of the nucleic acid sub-sequences used for creating error correction codes can vary, for example, they can be at least 3 nucleotides, at least 7 nucleotides, at least 15 nucleotides, or at least 31 nucleotides in length. In some embodiments, nucleic acid sub-sequences of other lengths can be used for creating error correction codes.


When a stochastic barcode comprises more than one of a type of label (e.g., more than one cell label or more than one molecular label), the labels may be interspersed with a linker label sequence. A linker label sequence can be at least about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length. A linker label sequence can be at most about 5, 10, 15, 20, 25, 30, 35, 40, 45, 50 or more nucleotides in length. In some instances, a linker label sequence is 12 nucleotides in length. A linker label sequence can be used to facilitate the synthesis of the stochastic barcode. The linker label can comprise an error-correcting (e.g., Hamming) code.


Solid Supports


The oligonucleotide probes (e.g., stochastic barcodes) and/or the blocking oligonucleotides disclosed herein can, in some embodiments, be attached to a solid support (e.g., bead, substrate, microwell(s), microwell arrays). As used herein, the terms “tethered”, “attached”, and “immobilized” are used interchangeably, and refer to covalent or non-covalent means for attaching a compound (e.g., an oligonucleotide) to a solid support. Any of a variety of different solid supports may be used as solid supports for attaching pre-synthesized combinatorial barcode reagents or for in situ solid-phase synthesis of combinatorial barcode reagents.


The solid support can be or comprise, for example, a particle or a plurality of particles. The particles can be, for example, nanoparticles, microparticles, or the likes. In some embodiments, a solid support is, or comprises, a bead or a plurality of beads. The particle (e.g., the bead) can encompass any type of solid, porous, or hollow sphere, ball, bearing, cylinder, or other similar configuration composed of plastic, ceramic, metal, or polymeric material onto which a nucleic acid may be immobilized (e.g., covalently or non-covalently). The particle (e.g., the bead) can comprise a discrete particle that may be spherical (e.g., microspheres) or have a non-spherical or irregular shape, such as cubic, cuboid, pyramidal, cylindrical, conical, oblong, or disc-shaped, and the like. The particle (e.g., the bead) can be spherical, substantial spherical, or non-spherical in shape.


The particle (e.g., bead) can comprise a variety of materials including, but not limited to, paramagnetic materials (e.g. magnesium, molybdenum, lithium, and tantalum), superparamagnetic materials (e.g. ferrite (Fe3O4; magnetite) nanoparticles), ferromagnetic materials (e.g. iron, nickel, cobalt, some alloys thereof, and some rare earth metal compounds), ceramic, plastic, glass, polystyrene, silica, methylstyrene, acrylic polymers, titanium, latex, sepharose, agarose, hydrogel, polymer, cellulose, nylon, and any combination thereof.


The diameter of the particle (e.g., the bead) can be, or be at least about, 5 μm, 10 μm, 20 μm, 25 μm, 30 μm, 35 μm, 40 μm, 45 μm, 50 μm, or in a range of any two of these values. The diameter of the particle (e.g., the bead) can be, for examples, at most about 5 μm, 10 μm, 20 μm, 25 μm, 30 μm, 35 μm, 40 μm, 45 μm or 50 μm. The diameter of the particle (e.g., the bead) can be related to the diameter of the wells of the substrate. For example, the diameter of the particle (e.g., bead) can be, or be at least, 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100% longer or shorter than the diameter of the well. In some embodiments, the diameter of the particle (e.g., bead) can be at most 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100% longer or shorter than the diameter of the well. The diameter of the particle (e.g., bead) can be related to the diameter of a cell (e.g., a single cell entrapped by a well of the substrate). The diameter of the particle (e.g., bead) can be, or be at least, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, or 300% or more longer or shorter than the diameter of the cell. In some embodiments, the diameter of the particle (e.g., bead) can be at most 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, or 300% or more longer or shorter than the diameter of the cell.


A particle (e.g., bead) can be attached to and/or embedded in a substrate. For example, the particle (e.g., bead) can be attached to and/or embedded in a gel, hydrogel, polymer and/or matrix. The spatial position of the particle (e.g., bead) within a substrate (e.g., gel, matrix, scaffold, or polymer) can be identified, in some embodiments, using the spatial label present on the stochastic barcode on the bead which can serve as a location address.


Examples of the particles (e.g., beads) can include, but are not limited to, streptavidin beads, agarose beads, magnetic beads, Dynabeads®, MACS® microbeads, antibody conjugated beads (e.g., anti-immunoglobulin microbead), protein A conjugated beads, protein G conjugated beads, protein A/G conjugated beads, protein L conjugated beads, oligodT conjugated beads, silica beads, silica-like beads, anti-biotin microbead, anti-fluorochrome microbead, and BcMag™ Carboxy-Terminated Magnetic Beads.


A particle (e.g., bead) can be associated with (e.g. impregnated with) quantum dots or fluorescent dyes to make it fluorescent in one fluorescence optical channel or multiple optical channels. A bead can be associated with iron oxide or chromium oxide to make it paramagnetic or ferromagnetic. The particles (e.g., beads) can be identifiable. A particle (e.g., bead) can be imaged using a camera. A particle (e.g., bead) can have a detectable code associated with the bead. For example, a bead can comprise an RFID tag. A bead can comprise any detectable tag (e.g., UPC code, electronic barcode, etched identifier). A particle (e.g., bead) can change size, for example due to swelling in an organic or inorganic solution. A bead can be hydrophobic or hydrophilic. A particle (e.g., bead) can be biocompatible.


A solid support (e.g., bead) can be visualized. The solid support can comprise a visualizing tag (e.g., fluorescent dye). A solid support (e.g., bead) can be etched with an identifier (e.g., a number). The identifier can be visualized through imaging the solid supports (e.g., beads).


A solid support (e.g., bead) can comprise an insoluble, semi-soluble, or insoluble material. A solid support can be referred to as “functionalized” when it includes a linker, a scaffold, a building block, or other reactive moiety attached thereto, whereas a solid support may be “nonfunctionalized” when it lack such a reactive moiety attached thereto. The solid support can be employed free in solution, such as in a microtiter well format; in a flow-through format, such as in a column; or in a dipstick.


The solid support can comprise a membrane, paper, plastic, coated surface, flat surface, glass, slide, chip, or any combination thereof. A solid support can take the form of resins, gels, microspheres, or other geometric configurations. A solid support can comprise silica chips, microparticles, nanoparticles, plates, arrays, capillaries, flat supports such as glass fiber filters, glass surfaces, metal surfaces (steel, gold silver, aluminum, silicon and copper), glass supports, plastic supports, silicon supports, chips, filters, membranes, microwell plates, slides, plastic materials including multiwell plates or membranes (e.g., formed of polyethylene, polypropylene, polyamide, polyvinylidenedifluoride), and/or wafers, combs, pins or needles (e.g., arrays of pins suitable for combinatorial synthesis or analysis) or beads in an array of pits or nanoliter wells of flat surfaces such as wafers (e.g., silicon wafers), wafers with pits with or without filter bottoms.


The solid support can comprise a polymer matrix (e.g., gel, hydrogel). The polymer matrix may be able to permeate intracellular space (e.g., around organelles). The polymer matrix may able to be pumped throughout the circulatory system.


A solid support can comprise, or be, a biological molecule. For example, a solid support can be a nucleic acid, a protein, an antibody, a histone, a cellular compartment, a lipid, a carbohydrate, and the like. Solid supports that are biological molecules can be amplified, translated, transcribed, degraded, and/or modified (e.g., pegylated, sumoylated, acetylated, methylated). A solid support that is a biological molecule can provide spatial and time information in addition to the spatial label that is attached to the biological molecule. For example, a biological molecule can comprise a first confirmation when unmodified, but can change to a second confirmation when modified. The different conformations can expose stochastic barcodes of the disclosure to targets. For example, a biological molecule can comprise stochastic barcodes that are unaccessible due to folding of the biological molecule. Upon modification of the biological molecule (e.g., acetylation), the biological molecule can change conformation to expose the stochastic labels. The timing of the modification can provide another time dimension to the method of stochastic barcoding of the disclosure.


In some embodiments, the biological molecule comprising combinatorial barcode reagents of the disclosure can be located in the cytoplasm of a cell. Upon activation, the biological molecule can move to the nucleus, whereupon stochastic barcoding can take place. In this way, modification of the biological molecule can encode additional space-time information for the targets identified by the stochastic barcodes.


A dimension label can provide information about space-time of a biological event (e.g., cell division). For example, a dimension label can be added to a first cell, where the first cell can divide generating a second daughter cell, the second daughter cell can comprise all, some or none of the dimension labels. The dimension labels can be activated in the original cell and the daughter cell. In this way, the dimension label can provide information about time of combinatorial barcoding in distinct spaces.


Substrates


As used herein, a substrate can refer to a type of solid support. A substrate can refer to a solid support that can comprise combinatorial barcode reagents of the disclosure. A substrate can comprise a plurality of microwells. A microwell can comprise a small reaction chamber of defined volume. A microwell can entrap one or more cells. A microwell can entrap only one cell. A microwell can entrap one or more solid supports. A microwell can entrap only one solid support. In some instances, a microwell entraps a single cell and a single solid support (e.g., bead). A microwell can comprise combinatorial barcode reagents of the disclosure.


The microwells of the array can be fabricated in a variety of shapes and sizes. Well geometries can include, but are not limited to, cylindrical, conical, hemispherical, rectangular, or polyhedral (e.g., three dimensional geometries comprised of several planar faces, for example, hexagonal columns, octagonal columns, inverted triangular pyramids, inverted square pyramids, inverted pentagonal pyramids, inverted hexagonal pyramids, or inverted truncated pyramids). The microwells can comprise a shape that combines two or more of these geometries. For example, a microwell can be partly cylindrical, with the remainder having the shape of an inverted cone. A microwell can include two side-by-side cylinders, one of larger diameter (e.g. that corresponds roughly to the diameter of the beads) than the other (e.g. that corresponds roughly to the diameter of the cells), that are connected by a vertical channel (that is, parallel to the cylinder axes) that extends the full length (depth) of the cylinders. The opening of the microwell can be at the upper surface of the substrate. The opening of the microwell can be at the lower surface of the substrate. The closed end (or bottom) of the microwell can be flat. The closed end (or bottom) of the microwell can have a curved surface (e.g., convex or concave). The shape and/or size of the microwell can be determined based on the types of cells or solid supports to be trapped within the microwells.


The portion of the substrate between the wells can have a topology. For example, the portion of the substrate between the wells can be rounded. The portion of the substrate between the wells can be pointed. The spacing portion of the substrate between the wells can be flat. The portion of the substrate between the wells may not be flat. In some instances, the portion of the substrate between wells is rounded. In other words, the portion of the substrate that does not comprise a well can have a curved surface. The curved surface can be fabricated such that the highest point (e.g., apex) of the curved surface may be at the furthest point between the edges of two or more wells (e.g., equidistant from the wells). The curved surface can be fabricated such that the start of the curved surface is at the edge of a first microwell and creates a parabola that ends at the end of a second microwell. This parabola can be extended in 2 dimensions to capture microwells nearby on the hexagonal grid of wells. The curved surface can be fabricated such that the surface between the wells is higher and/or curved than the plane of the opening of the well. The height of the curved surface can be, or be at least, 0.1, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, or 7 or more micrometers. In some embodiments, the height of the curved surface can be at most 0.1, 0.5, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, or 7 or more micrometers.


Microwell dimensions can be characterized in terms of the diameter and depth of the well. As used herein, the diameter of the microwell refers to the largest circle that can be inscribed within the planar cross-section of the microwell geometry. The diameter of the microwells can range from about 1-fold to about 10-fold the diameter of the cells or solid supports to be trapped within the microwells. The microwell diameter can be, or be at least, 1-fold, at least 1.5-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, or at least 10-fold the diameter of the cells or solid supports to be trapped within the microwells. In some embodiments, the microwell diameter can be at most 10-fold, at most 5-fold, at most 4-fold, at most 3-fold, at most 2-fold, at most 1.5-fold, or at most 1-fold the diameter of the cells or solid supports to be trapped within the microwells. The microwell diameter can be about 2.5-fold the diameter of the cells or solid supports to be trapped within the microwells.


The diameter of the microwells can be specified in terms of absolute dimensions. The diameter of the microwells can range from about 5 to about 60 micrometers. The microwell diameter can be, or be at least, 5 micrometers, at least 10 micrometers, at least 15 micrometers, at least 20 micrometers, at least 25 micrometers, at least 30 micrometers, at least 35 micrometers, at least 40 micrometers, at least 45 micrometers, at least 50 micrometers, or at least 60 micrometers. The microwell diameter can be at most 60 micrometers, at most 50 micrometers, at most 45 micrometers, at most 40 micrometers, at most 35 micrometers, at most 30 micrometers, at most 25 micrometers, at most 20 micrometers, at most 15 micrometers, at most 10 micrometers, or at most 5 micrometers. The microwell diameter can be about 30 micrometers.


The microwell depth may be chosen to provide efficient trapping of cells and solid supports. The microwell depth may be chosen to provide efficient exchange of assay buffers and other reagents contained within the wells. The ratio of diameter to height (i.e. aspect ratio) may be chosen such that once a cell and solid support settle inside a microwell, they will not be displaced by fluid motion above the microwell. The dimensions of the microwell may be chosen such that the microwell has sufficient space to accommodate a solid support and a cell of various sizes without being dislodged by fluid motion above the microwell. The depth of the microwells can range from about 1-fold to about 10-fold the diameter of the cells or solid supports to be trapped within the microwells. The microwell depth can be, or be at least, 1-fold, at least 1.5-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, or at least 10-fold the diameter of the cells or solid supports to be trapped within the microwells. The microwell depth can be at most 10-fold, at most 5-fold, at most 4-fold, at most 3-fold, at most 2-fold, at most 1.5-fold, or at most 1-fold the diameter of the cells or solid supports to be trapped within the microwells. The microwell depth can be about 2.5-fold the diameter of the cells or solid supports to be trapped within the microwells.


The depth of the microwells can be specified in terms of absolute dimensions. The depth of the microwells may range from about 10 to about 60 micrometers. The microwell depth can be, or be at least, 10 micrometers, at least 20 micrometers, at least 25 micrometers, at least 30 micrometers, at least 35 micrometers, at least 40 micrometers, at least 50 micrometers, or at least 60 micrometers. The microwell depth can be at most 60 micrometers, at most 50 micrometers, at most 40 micrometers, at most 35 micrometers, at most 30 micrometers, at most 25 micrometers, at most 20 micrometers, or at most 10 micrometers. The microwell depth can be about 30 micrometers.


The volume of the microwells used in the methods, devices, and systems of the present disclosure can range from about 200 micrometers3 to about 120,000 micrometers3. The microwell volume can be at least 200 micrometers3, at least 500 micrometers3, at least 1,000 micrometers3, at least 10,000 micrometers3, at least 25,000 micrometers3, at least 50,000 micrometers3, at least 100,000 micrometers3, or at least 120,000 micrometers3. The microwell volume can be at most 120,000 micrometers3, at most 100,000 micrometers3, at most 50,000 micrometers3, at most 25,000 micrometers3, at most 10,000 micrometers3, at most 1,000 micrometers3, at most 500 micrometers3, or at most 200 micrometers3. The microwell volume can be about 25,000 micrometers3. The microwell volume may fall within any range bounded by any of these values (e.g. from about 18,000 micrometers3 to about 30,000 micrometers3).


The volume of the microwell can be, or be at least, 5, 10, 15, 20, 25, 30, 35 40, 45 or 50 or more nanoliters3. The volume of the microwell can be at most 5, 10, 15, 20, 25, 30, 35 40, 45 or 50 or more nanoliters3. The volume of liquid that can fit in the microwell can be at least 5, 10, 15, 20, 25, 30, 35 40, 45 or 50 or more nanoliters3. The volume of liquid that can fit in the microwell can be at most 5, 10, 15, 20, 25, 30, 35 40, 45 or 50 or more nanoliters3. The volume of the microwell can be, or be at least, 5, 10, 15, 20, 25, 30, 35 40, 45 or 50 or more picoliters3. The volume of the microwell can be at most 5, 10, 15, 20, 25, 30, 35 40, 45 or 50 or more picoliters3. The volume of liquid that can fit in the microwell can be at least 5, 10, 15, 20, 25, 30, 35 40, 45 or 50 or more picoliters3. The volume of liquid that can fit in the microwell can be at most 5, 10, 15, 20, 25, 30, 35 40, 45 or 50 or more picoliters3.


The volumes of the microwells used in the methods, devices, and systems of the present disclosure may be further characterized in terms of the variation in volume from one microwell to another. The coefficient of variation (expressed as a percentage) for microwell volume may range from about 1% to about 10%. The coefficient of variation for microwell volume may be at least 1%, at least 2%, at least 3%, at least 4%, at least 5%, at least 6%, at least 7%, at least 8%, at least 9%, or at least 10%. The coefficient of variation for microwell volume may be at most 10%, at most 9%, at most 8%, at most 7%, at most 6%, at most 5%, at most 4%, at most 3%, at most 2%, or at most 1%. The coefficient of variation for microwell volume may have any value within a range encompassed by these values, for example between about 1.5% and about 6.5%. In some embodiments, the coefficient of variation of microwell volume may be about 2.5%.


The ratio of the volume of the microwells to the surface area of the beads (or to the surface area of a solid support to which stochastic barcode oligonucleotides may be attached) used in the methods, devices, and systems of the present disclosure can range from about 2.5 to about 1,520 micrometers. The ratio can be at least 2.5, at least 5, at least 10, at least 100, at least 500, at least 750, at least 1,000, or at least 1,520. The ratio can be at most 1,520, at most 1,000, at most 750, at most 500, at most 100, at most 10, at most 5, or at most 2.5. The ratio can be about 67.5. The ratio of microwell volume to the surface area of the bead (or solid support used for immobilization) may fall within any range bounded by any of these values (e.g. from about 30 to about 120).


The wells of the microwell array can be arranged in a one dimensional, two dimensional, or three-dimensional array. In some embodiments, a three dimensional array can be achieved, for example, by stacking a series of two or more two dimensional arrays (that is, by stacking two or more substrates comprising microwell arrays).


The pattern and spacing between microwells can be chosen to optimize the efficiency of trapping a single cell and single solid support (e.g., bead) in each well, as well as to maximize the number of wells per unit area of the array. The microwells may be distributed according to a variety of random or non-random patterns. For example, they may be distributed entirely randomly across the surface of the array substrate, or they may be arranged in a square grid, rectangular grid, hexagonal grid, or the like. In some instances, the microwells are arranged hexagonally. The center-to-center distance (or spacing) between wells may vary from about 5 micrometers to about 75 micrometers. In some instances, the spacing between microwells is about 10 micrometers. In other embodiments, the spacing between wells is at least 5 micrometers, at least 10 micrometers, at least 15 micrometers, at least 20 micrometers, at least 25 micrometers, at least 30 micrometers, at least 35 micrometers, at least 40 micrometers, at least 45 micrometers, at least 50 micrometers, at least 55 micrometers, at least 60 micrometers, at least 65 micrometers, at least 70 micrometers, or at least 75 micrometers. The microwell spacing can be at most 75 micrometers, at most 70 micrometers, at most 65 micrometers, at most 60 micrometers, at most 55 micrometers, at most 50 micrometers, at most 45 micrometers, at most 40 micrometers, at most 35 micrometers, at most 30 micrometers, at most 25 micrometers, at most 20 micrometers, at most 15 micrometers, at most 10 micrometers, at most 5 micrometers. The microwell spacing can be about 55 micrometers. The microwell spacing may fall within any range bounded by any of these values (e.g. from about 18 micrometers to about 72 micrometers).


The microwell array may comprise surface features between the microwells that are designed to help guide cells and solid supports into the wells and/or prevent them from settling on the surfaces between wells. Examples of suitable surface features can include, but are not limited to, domed, ridged, or peaked surface features that encircle the wells or straddle the surface between wells.


The total number of wells in the microwell array can be determined by the pattern and spacing of the wells and the overall dimensions of the array. The number of microwells in the array can range from about 96 to about 5,000,000 or more. The number of microwells in the array can be at least 96, at least 384, at least 1,536, at least 5,000, at least 10,000, at least 25,000, at least 50,000, at least 75,000, at least 100,000, at least 500,000, at least 1,000,000, or at least 5,000,000. The number of microwells in the array can be at most 5,000,000, at most 1,000,000, at most 75,000, at most 50,000, at most 25,000, at most 10,000, at most 5,000, at most 1,536, at most 384, or at most 96 wells. The number of microwells in the array can be about 96, 384, and/or 1536. The number of microwells can be about 150,000. The number of microwells in the array may fall within any range bounded by any of these values (e.g. from about 100 to 325,000).


Microwell arrays may be fabricated using any of a number of fabrication techniques. Examples of fabrication methods that may be used include, but are not limited to, bulk micromachining techniques such as photolithography and wet chemical etching, plasma etching, or deep reactive ion etching; micro-molding and micro-embossing; laser micromachining; 3D printing or other direct write fabrication processes using curable materials; and similar techniques.


Microwell arrays can be fabricated from any of a number of substrate materials. The choice of material can depend on the choice of fabrication technique, and vice versa. Examples of suitable materials can include, but are not limited to, silicon, fused-silica, glass, polymers (e.g. agarose, gelatin, hydrogels, polydimethylsiloxane (PDMS; elastomer), polymethylmethacrylate (PMMA), polycarbonate (PC), polypropylene (PP), polyethylene (PE), high density polyethylene (HDPE), polyimide, cyclic olefin polymers (COP), cyclic olefin copolymers (COC), polyethylene terephthalate (PET), epoxy resins, thiol-ene based resins, metals or metal films (e.g. aluminum, stainless steel, copper, nickel, chromium, and titanium), and the like. In some instances, the microwell comprises optical adhesive. In some instances, the microwell is made out of optical adhesive. In some instances, the microwell array comprises and/or is made out of PDMS. In some instances, the microwell is made of plastic. A hydrophilic material can be desirable for fabrication of the microwell arrays (e.g. to enhance wettability and minimize non-specific binding of cells and other biological material). Hydrophobic materials that can be treated or coated (e.g. by oxygen plasma treatment, or grafting of a polyethylene oxide surface layer) can also be used. The use of porous, hydrophilic materials for the fabrication of the microwell array may be desirable in order to facilitate capillary wicking/venting of entrapped air bubbles in the device. The microwell array can be fabricated from a single material. The microwell array may comprise two or more different materials that have been bonded together or mechanically joined.


Microwell arrays can be fabricated using substrates of any of a variety of sizes and shapes. For example, the shape (or footprint) of the substrate within which microwells are fabricated may be square, rectangular, circular, or irregular in shape. The footprint of the microwell array substrate can be similar to that of a microtiter plate. The footprint of the microwell array substrate can be similar to that of standard microscope slides, e.g. about 75 mm long×25 mm wide (about 3″ long×1″ wide), or about 75 mm long×50 mm wide (about 3″ long×2″ wide). The thickness of the substrate within which the microwells are fabricated may range from about 0.1 mm thick to about 10 mm thick, or more. The thickness of the microwell array substrate may be at least 0.1 mm thick, at least 0.5 mm thick, at least 1 mm thick, at least 2 mm thick, at least 3 mm thick, at least 4 mm thick, at least 5 mm thick, at least 6 mm thick, at least 7 mm thick, at least 8 mm thick, at least 9 mm thick, or at least 10 mm thick. The thickness of the microwell array substrate may be at most 10 mm thick, at most 9 mm thick, at most 8 mm thick, at most 7 mm thick, at most 6 mm thick, at most 5 mm thick, at most 4 mm thick, at most 3 mm thick, at most 2 mm thick, at most 1 mm thick, at most 0.5 mm thick, or at most 0.1 mm thick. The thickness of the microwell array substrate can be about 1 mm thick. The thickness of the microwell array substrate may be any value within these ranges, for example, the thickness of the microwell array substrate may be between about 0.2 mm and about 9.5 mm. The thickness of the microwell array substrate may be uniform.


A variety of surface treatments and surface modification techniques may be used to alter the properties of microwell array surfaces. Examples can include, but are not limited to, oxygen plasma treatments to render hydrophobic material surfaces more hydrophilic, the use of wet or dry etching techniques to smooth (or roughen) glass and silicon surfaces, adsorption or grafting of polyethylene oxide or other polymer layers (such as pluronic), or bovine serum albumin to substrate surfaces to render them more hydrophilic and less prone to non-specific adsorption of biomolecules and cells, the use of silane reactions to graft chemically-reactive functional groups to otherwise inert silicon and glass surfaces, etc. Photodeprotection techniques can be used to selectively activate chemically-reactive functional groups at specific locations in the array structure, for example, the selective addition or activation of chemically-reactive functional groups such as primary amines or carboxyl groups on the inner walls of the microwells may be used to covalently couple oligonucleotide probes, peptides, proteins, or other biomolecules to the walls of the microwells. The choice of surface treatment or surface modification utilized can depend both or either on the type of surface property that is desired and on the type of material from which the microwell array is made.


The openings of microwells can be sealed, for example, during cell lysis steps to prevent cross hybridization of target nucleic acid between adjacent microwells. A microwell (or array of microwells) may be sealed or capped using, for example, a flexible membrane or sheet of solid material (i.e. a plate or platten) that clamps against the surface of the microwell array substrate, or a suitable bead, where the diameter of the bead is larger than the diameter of the microwell.


A seal formed using a flexible membrane or sheet of solid material can comprise, for example, inorganic nanopore membranes (e.g., aluminum oxides), dialysis membranes, glass slides, coverslips, elastomeric films (e.g. PDMS), or hydrophilic polymer films (e.g., a polymer film coated with a thin film of agarose that has been hydrated with lysis buffer).


Solid supports (e.g., beads) used for capping the microwells may comprise any of the solid supports (e.g., beads) of the disclosure. In some instances, the solid supports are cross-linked dextran beads (e.g., Sephadex). Cross-linked dextran can range from about 10 micrometers to about 80 micrometers. The cross-linked dextran beads used for capping can be from 20 micrometers to about 50 micrometers. In some embodiments, the beads may be at least about 10, 20, 30, 40, 50, 60, 70, 80 or 90% larger than the diameter of the microwells. The beads used for capping may be at most about 10, 20, 30, 40, 50, 60, 70, 80 or 90% larger than the diameter of the microwells.


The seal or cap may allow buffer to pass into and out of the microwell, while preventing macromolecules (e.g., nucleic acids) from migrating out of the well. A macromolecule of at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, or 20 or more nucleotides may be blocked from migrating into or out of the microwell by the seal or cap. A macromolecule of at most about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 13, 14, 15, 16, 17, 18, 19, or 20 or more nucleotides may be blocked from migrating into or out of the microwell by the seal or cap.


Solid supports (e.g., beads) may be distributed among a substrate. Solid supports (e.g., beads) can be distributed among wells of the substrate, removed from the wells of the substrate, or otherwise transported through a device comprising one or more microwell arrays by means of centrifugation or other non-magnetic means. A microwell of a substrate can be pre-loaded with a solid support. A microwell of a substrate can hold at least 1, 2, 3, 4, or 5, or more solid supports. A microwell of a substrate can hold at most 1, 2, 3, 4, or 5 or more solid supports. In some instances, a microwell of a substrate can hold one solid support.


Individual cells and beads may be compartmentalized using alternatives to microwells, for example, a single solid support and single cell could be confined within a single droplet in an emulsion (e.g. in a droplet digital microfluidic system).


Cells could potentially be confined within porous beads that themselves comprise the plurality of tethered stochastic barcodes. Individual cells and solid supports may be compartmentalized in any type of container, microcontainer, reaction chamber, reaction vessel, or the like.


Single cell combinatorial barcoding or may be performed without the use of microwells. Single cell, combinatorial barcoding assays may be performed without the use of any physical container. For example, combinatorial barcoding without a physical container can be performed by embedding cells and beads in close proximity to each other within a polymer layer or gel layer to create a diffusional barrier between different cell/bead pairs. In another example, combinatorial barcoding without a physical container can be performed in situ, in vivo, on an intact solid tissue, on an intact cell, and/or subcellularly.


Microwell arrays can be a consumable component of the assay system. Microwell arrays may be reusable. Microwell arrays can be configured for use as a stand-alone device for performing assays manually, or they may be configured to comprise a fixed or removable component of an instrument system that provides for full or partial automation of the assay procedure. In some embodiments of the disclosed methods, the bead-based libraries of stochastic barcodes can be deposited in the wells of the microwell array as part of the assay procedure. In some embodiments, the beads may be pre-loaded into the wells of the microwell array and provided to the user as part of, for example, a kit for performing stochastic barcoding and digital counting of nucleic acid targets.


In some embodiments, two mated microwell arrays are provided, one pre-loaded with beads which are held in place by a first magnet, and the other for use by the user in loading individual cells. Following distribution of cells into the second microwell array, the two arrays may be placed face-to-face and the first magnet removed while a second magnet is used to draw the beads from the first array down into the corresponding microwells of the second array, thereby ensuring that the beads rest above the cells in the second microwell array and thus minimizing diffusional loss of target molecules following cell lysis, while maximizing efficient attachment of target molecules to the stochastic barcodes on the bead.


Microwell arrays of the disclosure can be pre-loaded with solid supports (e.g., beads). Each well of a microwell array can comprise a single solid support. At least 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% of the wells in a microwell array can be pre-loaded with a single solid support. At most 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% of the wells in a microwell array can be pre-loaded with a single solid support. The solid support can comprise stochastic barcodes and/or combinatorial barcodes of the disclosure. Cell labels of stochastic barcodes on different solid supports can be different. Cell labels of stochastic barcodes on the same solid support can be the same.


While various aspects and embodiments have been disclosed herein, other aspects and embodiments will be apparent to those skilled in the art. The various aspects and embodiments disclosed herein are for purposes of illustration and are not intended to be limiting, with the true scope and spirit being indicated by the following claims.


One skilled in the art will appreciate that, for this and other processes and methods disclosed herein, the functions performed in the processes and methods can be implemented in differing order. Furthermore, the outlined steps and operations are only provided as examples, and some of the steps and operations can be optional, combined into fewer steps and operations, or expanded into additional steps and operations without detracting from the essence of the disclosed embodiments.


With respect to the use of substantially any plural and/or singular terms herein, those having skill in the art can translate from the plural to the singular and/or from the singular to the plural as is appropriate to the context and/or application. The various singular/plural permutations may be expressly set forth herein for sake of clarity.


It will be understood by those within the art that, in general, terms used herein, and especially in the appended claims (e.g., bodies of the appended claims) are generally intended as “open” terms (e.g., the term “including” should be interpreted as “including but not limited to,” the term “having” should be interpreted as “having at least,” the term “includes” should be interpreted as “includes but is not limited to,” etc.). It will be further understood by those within the art that if a specific number of an introduced claim recitation is intended, such an intent will be explicitly recited in the claim, and in the absence of such recitation no such intent is present. For example, as an aid to understanding, the following appended claims may contain usage of the introductory phrases “at least one” and “one or more” to introduce claim recitations. However, the use of such phrases should not be construed to imply that the introduction of a claim recitation by the indefinite articles “a” or “an” limits any particular claim containing such introduced claim recitation to embodiments containing only one such recitation, even when the same claim includes the introductory phrases “one or more” or “at least one” and indefinite articles such as “a” or “an” (e.g., “a” and/or “an” should be interpreted to mean “at least one” or “one or more”); the same holds true for the use of definite articles used to introduce claim recitations. In addition, even if a specific number of an introduced claim recitation is explicitly recited, those skilled in the art will recognize that such recitation should be interpreted to mean at least the recited number (e.g., the bare recitation of “two recitations,” without other modifiers, means at least two recitations, or two or more recitations). Furthermore, in those instances where a convention analogous to “at least one of A, B, and C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., “a system having at least one of A, B, and C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). In those instances where a convention analogous to “at least one of A, B, or C, etc.” is used, in general such a construction is intended in the sense one having skill in the art would understand the convention (e.g., “a system having at least one of A, B, or C” would include but not be limited to systems that have A alone, B alone, C alone, A and B together, A and C together, B and C together, and/or A, B, and C together, etc.). It will be further understood by those within the art that virtually any disjunctive word and/or phrase presenting two or more alternative terms, whether in the description, claims, or drawings, should be understood to contemplate the possibilities of including one of the terms, either of the terms, or both terms. For example, the phrase “A or B” will be understood to include the possibilities of “A” or “B” or “A and B.”


In addition, where features or aspects of the disclosure are described in terms of Markush groups, those skilled in the art will recognize that the disclosure is also thereby described in terms of any individual member or subgroup of members of the Markush group.


As will be understood by one skilled in the art, for any and all purposes, such as in terms of providing a written description, all ranges disclosed herein also encompass any and all possible subranges and combinations of subranges thereof. Any listed range can be easily recognized as sufficiently describing and enabling the same range being broken down into at least equal halves, thirds, quarters, fifths, tenths, etc. As a non-limiting example, each range discussed herein can be readily broken down into a lower third, middle third and upper third, etc. As will also be understood by one skilled in the art all language such as “up to,” “at least,” and the like include the number recited and refer to ranges which can be subsequently broken down into subranges as discussed above. Finally, as will be understood by one skilled in the art, a range includes each individual member. Thus, for example, a group having 1-3 cells refers to groups having 1, 2, or 3 cells. Similarly, a group having 1-5 cells refers to groups having 1, 2, 3, 4, or 5 cells, and so forth.


From the foregoing, it will be appreciated that various embodiments of the present disclosure have been described herein for purposes of illustration, and that various modifications may be made without departing from the scope and spirit of the present disclosure. Accordingly, the various embodiments disclosed herein are not intended to be limiting, with the true scope and spirit being indicated by the following claims.

Claims
  • 1. A method of selective amplification, comprising: providing a sample comprising a plurality of nucleic acid target molecules and one or more undesirable nucleic acid species;providing a plurality of particles each comprising a plurality of oligonucleotide probes, wherein each of the plurality of oligonucleotide probes comprises a molecular label sequence, a cell label sequence and a binding region, wherein the plurality of oligonucleotide probes comprises at least 100 different molecular label sequences, and wherein oligonucleotide probes associated with the same particle comprise the same cell label sequence and oligonucleotide probes associated with different particles comprise different cell label sequences;contacting the plurality of oligonucleotide probes with the plurality of nucleic acid target molecules for hybridization;extending oligonucleotide probes that are hybridized to the plurality of nucleic acid target molecules to generate a plurality of extension products;providing one or more amplification primers;providing a blocking oligonucleotide that specifically binds to at least one of the one or more undesirable nucleic acid species within 100 nt of the 5′ end of the one or more undesirable nucleic acid species; andamplifying the plurality of extension products in the presence of the blocking oligonucleotide and one or more amplification primers to generate a plurality of amplicons, wherein the one or more amplification primers add sequencing adaptors to the plurality of extension products,whereby the amplification of the undesirable nucleic acid species is reduced by the blocking oligonucleotide not being able to function as a primer for a polymerase.
  • 2. The method of claim 1, wherein the blocking oligonucleotide comprises a locked nucleic acid (LNA), a peptide nucleic acid (PNA), a DNA, an LNA/PNA chimera, an LNA/DNA chimera, a PNA/DNA chimera, or any combination thereof.
  • 3. The method of claim 1, comprising providing blocking oligonucleotides that specifically bind to two or more, at least 10, or at least 100 undesirable nucleic acid species in the sample.
  • 4. The method of claim 1, wherein the blocking oligonucleotide has a Tm of at least 60° C., 65° C., or 70° C.
  • 5. The method of claim 1, wherein the one or more undesirable nucleic acid species amounts to about 50%, 60%, 70%, or 80% of the nucleic acid content of the sample.
  • 6. The method of claim 1, wherein the undesirable nucleic acid species comprise rRNA, mtRNA, genomic DNA, intronic sequence, high-abundance sequence, or any combination thereof.
  • 7. The method of claim 1, wherein the blocking oligonucleotide specifically binds to within 50 nt of the 5′ end of the one or more undesirable nucleic acid species.
  • 8. The method of claim 1, wherein the amplifying comprises PCR amplification of the plurality of extension products.
  • 9. The method of claim 1, wherein the plurality of oligonucleotide probes comprises at least 1000 or 10000 different molecular label sequences.
  • 10. The method of claim 1, wherein the plurality of amplicons comprises a cDNA library.
  • 11. The method of claim 1, wherein the sample comprises a single cell, a plurality of cells, or a tissue sample.
  • 12. The method of claim 1, further comprising sequencing the plurality of amplicons.
  • 13. The method of claim 1, wherein the plurality of oligonucleotide probes is immobilized on the particle.
  • 14. The method of claim 1, wherein the plurality of nucleic acid target molecules comprises mRNA target molecules.
  • 15. The method of claim 1, wherein the binding region comprises a poly-dT sequence.
  • 16. The method of claim 1, wherein the plurality of particles comprises streptavidin beads, agarose beads, magnetic beads, protein A-conjugated beads, protein G-conjugated beads, protein A/G-conjugated beads, protein L-conjugated beads, oligo(dT)-conjugated beads, silica beads, anti-biotin microbeads, or anti-fluorochrome microbeads.
  • 17. The method of claim 1, wherein the blocking oligonucleotide specifically binds to within 10 nt of the 5′ end of the one or more undesirable nucleic acid species.
  • 18. A method of selective extension, comprising: providing a sample comprising a plurality of nucleic acid target molecules and one or more undesirable nucleic acid species;providing a plurality of particles each comprising a plurality of oligonucleotide probes, wherein each of the plurality of oligonucleotide probes comprises a molecular label sequence, a cell label sequence and a binding region, wherein the plurality of oligonucleotide probes comprises at least 100 different molecular label sequences, and wherein oligonucleotide probes associated with the same particle comprise the same cell label sequence and oligonucleotide probes associated with different particles comprise different cell label sequences;contacting the plurality of oligonucleotide probes with the plurality of nucleic acid target molecules for hybridization;providing a blocking oligonucleotide that specifically binds to at least one of the one or more undesirable nucleic acid species within 100 nt of the 5′ end of the one or more undesirable nucleic acid species; andextending oligonucleotide probes that are hybridized to the plurality of nucleic acid target molecules in the presence of the blocking oligonucleotide to generate a plurality of extension products;whereby the extension of oligonucleotide probes hybridized to the undesirable nucleic acid species is reduced by the blocking oligonucleotide not being able to function as a primer for a polymerase.
  • 19. The method of claim 18, wherein the blocking oligonucleotide comprises a locked nucleic acid (LNA), a peptide nucleic acid (PNA), a DNA, an LNA/PNA chimera, an LNA/DNA chimera, a PNA/DNA chimera, or any combination thereof.
  • 20. The method of claim 18, wherein the plurality of particles comprises streptavidin beads, agarose beads, magnetic beads, protein A-conjugated beads, protein G-conjugated beads, protein A/G-conjugated beads, protein L-conjugated beads, oligo(dT)-conjugated beads, silica beads, anti-biotin microbeads, or anti-fluorochrome microbeads.
  • 21. The method of claim 18, wherein the blocking oligonucleotide specifically binds to within 50 nt of the 5′ end of the one or more undesirable nucleic acid species.
  • 22. The method of claim 18, wherein the blocking oligonucleotide specifically binds to within 10 nt of the 5′ end of the one or more undesirable nucleic acid species.
  • 23. The method of claim 18, further comprising sequencing the plurality of extension products, or products thereof, to generate a plurality of sequencing reads.
RELATED APPLICATIONS

The present application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Application No. 62/453,163, filed on Feb. 1, 2017. The content of this related application is expressly incorporated herein by reference in its entirety.

US Referenced Citations (557)
Number Name Date Kind
4510244 Parks et al. Apr 1985 A
4725536 Fritsch et al. Feb 1988 A
5124246 Urdea et al. Jun 1992 A
5149625 Church et al. Sep 1992 A
5200314 Urdea Apr 1993 A
5308990 Takahashi et al. May 1994 A
5424186 Fodor et al. Jun 1995 A
5424413 Hogan et al. Jun 1995 A
5445934 Fodor et al. Aug 1995 A
5604097 Brenner Feb 1997 A
5635352 Urdea et al. Jun 1997 A
5635400 Brenner Jun 1997 A
5648245 Fire et al. Jul 1997 A
5654413 Brenner Aug 1997 A
5656731 Urdea Aug 1997 A
5658737 Nelson et al. Aug 1997 A
5714330 Brenner et al. Feb 1998 A
5744305 Fodor et al. Apr 1998 A
5759778 Li et al. Jun 1998 A
5763175 Brenner Jun 1998 A
5800992 Fodor et al. Sep 1998 A
5830712 Rampersad et al. Nov 1998 A
5846719 Brenner et al. Dec 1998 A
5854033 Lizardi Dec 1998 A
5871928 Fodor et al. Feb 1999 A
5925525 Fodor et al. Jul 1999 A
5935793 Wong Aug 1999 A
5962271 Chenchik et al. Oct 1999 A
5962272 Chenchik et al. Oct 1999 A
5968740 Fodor et al. Oct 1999 A
5981176 Wallace Nov 1999 A
5981179 Lorinez et al. Nov 1999 A
6013445 Albrecht et al. Jan 2000 A
6040138 Lockhart et al. Mar 2000 A
6046005 Ju et al. Apr 2000 A
6060596 Lerner et al. May 2000 A
6064755 Some May 2000 A
6114149 Fry et al. Sep 2000 A
6117631 Nilsen Sep 2000 A
6124092 O'neill et al. Sep 2000 A
6138077 Brenner Oct 2000 A
6140489 Brenner Oct 2000 A
6172214 Brenner Jan 2001 B1
6197506 Fodor et al. Mar 2001 B1
6197554 Lin et al. Mar 2001 B1
6214558 Shuber et al. Apr 2001 B1
6235475 Brenner et al. May 2001 B1
6235483 Wolber et al. May 2001 B1
6265163 Albrecht et al. Jul 2001 B1
6268152 Fodor et al. Jul 2001 B1
6284460 Fodor et al. Sep 2001 B1
6284485 Boyle et al. Sep 2001 B1
6309822 Fodor et al. Oct 2001 B1
6309823 Cronin et al. Oct 2001 B1
6326148 Pauletti et al. Dec 2001 B1
6355431 Chee et al. Mar 2002 B1
6355432 Fodor et al. Mar 2002 B1
6372813 Johnson et al. Apr 2002 B1
6395491 Fodor et al. May 2002 B1
6406848 Bridgham et al. Jun 2002 B1
6436675 Welch et al. Aug 2002 B1
6440667 Fodor et al. Aug 2002 B1
6440706 Vogelstein et al. Aug 2002 B1
6451536 Fodor et al. Sep 2002 B1
6458530 Morris et al. Oct 2002 B1
6468744 Cronin et al. Oct 2002 B1
6480791 Strathmann Nov 2002 B1
6489114 Laayoun et al. Dec 2002 B2
6489116 Wagner Dec 2002 B2
6492121 Kurane et al. Dec 2002 B2
6500620 Yu et al. Dec 2002 B2
6512105 Hogan et al. Jan 2003 B1
6514699 O'neill et al. Feb 2003 B1
6544739 Fodor et al. Apr 2003 B1
6551784 Fodor et al. Apr 2003 B2
6576424 Fodor et al. Jun 2003 B2
6600996 Webster et al. Jul 2003 B2
6629040 Goodlett et al. Sep 2003 B1
6653077 Brenner Nov 2003 B1
6753147 Vogelstein et al. Jun 2004 B2
6787308 Balasubramanian et al. Sep 2004 B2
6808906 Shen et al. Oct 2004 B2
6849404 Park et al. Feb 2005 B2
6852488 Fodor et al. Feb 2005 B2
6858412 Willis et al. Feb 2005 B2
6890741 Fan et al. May 2005 B2
6946251 Kurn Sep 2005 B2
6974669 Mirkin et al. Dec 2005 B2
7022479 Wagner Apr 2006 B2
7034145 Shen et al. Apr 2006 B2
7155050 Sloge Dec 2006 B1
7294466 McMillan Nov 2007 B2
7323309 Mirkin et al. Jan 2008 B2
7393665 Brenner Jul 2008 B2
7407757 Brenner Aug 2008 B2
7424368 Huang et al. Sep 2008 B2
7432055 Pemov et al. Oct 2008 B2
7470515 Rashtchian et al. Dec 2008 B2
7473767 Dimitrov Jan 2009 B2
7476786 Chan et al. Jan 2009 B2
7537897 Brenner et al. May 2009 B2
7544473 Brenner Jun 2009 B2
7635566 Brenner Dec 2009 B2
7718403 Kamberov et al. May 2010 B2
7771946 Kurn Aug 2010 B2
7822555 Huang et al. Oct 2010 B2
7824856 Monforte Nov 2010 B2
7824889 Vogelstein et al. Nov 2010 B2
7915015 Vogelstein et al. Mar 2011 B2
7985546 Church et al. Jul 2011 B2
8071311 Kurn Dec 2011 B2
8110351 Bosnes Feb 2012 B2
8114681 Martin et al. Feb 2012 B2
8148068 Brenner Apr 2012 B2
8168385 Brenner May 2012 B2
8206913 Kamberov et al. Jun 2012 B1
8241850 Brenner Aug 2012 B2
8298767 Brenner et al. Oct 2012 B2
8318433 Brenner Nov 2012 B2
8367051 Matyjaszewski et al. Feb 2013 B2
8420324 Rashtchian et al. Apr 2013 B2
8445205 Brenner May 2013 B2
8470996 Brenner Jun 2013 B2
8476018 Brenner Jul 2013 B2
8481292 Casbon et al. Jul 2013 B2
8535889 Larson et al. Sep 2013 B2
8563274 Brenner et al. Oct 2013 B2
8603749 Gillevet Dec 2013 B2
8679756 Brenner et al. Mar 2014 B1
8685678 Casbon et al. Apr 2014 B2
8685753 Martin et al. Apr 2014 B2
8715967 Casbon et al. May 2014 B2
8722368 Casbon et al. May 2014 B2
8728766 Casbon et al. May 2014 B2
8741606 Casbon et al. Jun 2014 B2
8835110 Wang et al. Sep 2014 B2
8835358 Fodor et al. Sep 2014 B2
8841071 Link Sep 2014 B2
8856410 Park Oct 2014 B2
9150852 Samuels et al. Oct 2015 B2
9181582 Kurn Nov 2015 B2
9228229 Olson et al. Jan 2016 B2
9262376 Tsuto Feb 2016 B2
9290808 Fodor et al. Mar 2016 B2
9290809 Fodor et al. Mar 2016 B2
9297047 Furchak et al. Mar 2016 B2
9315857 Fu et al. Apr 2016 B2
9371598 Chee Jun 2016 B2
9567645 Fan et al. Feb 2017 B2
9567646 Fan et al. Feb 2017 B2
9582877 Fu et al. Feb 2017 B2
9593365 Frisen et al. Mar 2017 B2
9598736 Fan et al. Mar 2017 B2
9637799 Fan et al. May 2017 B2
9644204 Hindson et al. May 2017 B2
9689024 Hindson et al. Jun 2017 B2
9695468 Hindson et al. Jul 2017 B2
9708659 Fodor et al. Jul 2017 B2
9727810 Fodor et al. Aug 2017 B2
9787810 Chiang Oct 2017 B1
9816137 Fodor et al. Nov 2017 B2
9845502 Fodor et al. Dec 2017 B2
9850515 McCoy et al. Dec 2017 B2
9856530 Hindson et al. Jan 2018 B2
9868979 Chee et al. Jan 2018 B2
9879313 Chee et al. Jan 2018 B2
9905005 Fu et al. Feb 2018 B2
9938523 LaBaer Apr 2018 B2
9951386 Hindson et al. Apr 2018 B2
9988660 Rashtchian et al. Jun 2018 B2
10002316 Fodor et al. Jun 2018 B2
10011872 Belgrader et al. Jul 2018 B1
10017761 Weissman et al. Jul 2018 B2
10023910 Drmanac et al. Jul 2018 B2
10030267 Hindson et al. Jul 2018 B2
10041116 Hindson et al. Aug 2018 B2
10047394 Fodor et al. Aug 2018 B2
10059991 Fodor et al. Aug 2018 B2
10131958 Fan et al. Nov 2018 B1
10138518 Chun Nov 2018 B2
10151003 Fan et al. Dec 2018 B2
10202641 Shum Feb 2019 B2
10202646 Fodor et al. Feb 2019 B2
10208343 Hindson et al. Feb 2019 B2
10208356 Fan et al. Feb 2019 B1
10227648 Hindson et al. Mar 2019 B2
10246703 Church et al. Apr 2019 B2
10253364 Hindson et al. Apr 2019 B2
10253375 Fan et al. Apr 2019 B1
10273541 Hindson et al. Apr 2019 B2
10288608 Kozlov et al. May 2019 B2
10294511 Sanches-Kuiper et al. May 2019 B2
10308982 Chee Jun 2019 B2
10323278 Belgrader et al. Jun 2019 B2
10337061 Hindson et al. Jul 2019 B2
10338066 Fan et al. Jul 2019 B2
10344329 Hindson et al. Jul 2019 B2
10392661 Fodor et al. Aug 2019 B2
10450607 Hindson et al. Oct 2019 B2
10550429 Harada et al. Feb 2020 B2
10619186 Betts et al. Apr 2020 B2
10619203 Fodor et al. Apr 2020 B2
11092607 Gaublomme et al. Aug 2021 B2
20010024784 Wagner Sep 2001 A1
20010036632 Yu et al. Nov 2001 A1
20020019005 Kamb Feb 2002 A1
20020051986 Baez et al. May 2002 A1
20020065609 Ashby May 2002 A1
20020072058 Voelker et al. Jun 2002 A1
20020094116 Forst et al. Jul 2002 A1
20020106666 Hayashizaki Aug 2002 A1
20020132241 Fan et al. Sep 2002 A1
20020168665 Okawa Nov 2002 A1
20020172953 Mirkin et al. Nov 2002 A1
20020187480 Brandon Dec 2002 A1
20020192687 Mirkin et al. Dec 2002 A1
20030003490 Fan et al. Jan 2003 A1
20030013091 Dimitrov Jan 2003 A1
20030032049 Wagner Feb 2003 A1
20030049616 Brenner et al. Mar 2003 A1
20030077611 Slepnev Apr 2003 A1
20030082818 Bahnson et al. May 2003 A1
20030087242 Mirkin et al. May 2003 A1
20030104436 Morris et al. Jun 2003 A1
20030165935 Vann et al. Sep 2003 A1
20030175908 Linnarson Sep 2003 A1
20030186251 Dunn et al. Oct 2003 A1
20030207296 Park et al. Nov 2003 A1
20030207300 Matray et al. Nov 2003 A1
20040047769 Tanaami Mar 2004 A1
20040091864 French et al. May 2004 A1
20040096368 Davis et al. May 2004 A1
20040096892 Wang et al. May 2004 A1
20040121342 McKeown Jun 2004 A1
20040146901 Morris et al. Jul 2004 A1
20040147435 Hawiger et al. Jul 2004 A1
20040157243 Huang et al. Aug 2004 A1
20040180361 Dahl Sep 2004 A1
20040224325 Knapp et al. Nov 2004 A1
20040259118 Macevicz Dec 2004 A1
20050019776 Callow et al. Jan 2005 A1
20050032110 Shen et al. Feb 2005 A1
20050048500 Lawton Mar 2005 A1
20050053952 Hong et al. Mar 2005 A1
20050105077 Padmanabhan et al. May 2005 A1
20050170373 Monforte Aug 2005 A1
20050175993 Wei Aug 2005 A1
20050196760 Pemov et al. Sep 2005 A1
20050214825 Stuelpnagel Sep 2005 A1
20050250146 McMillan Nov 2005 A1
20050250147 Macevicz Nov 2005 A1
20060002824 Chang et al. Jan 2006 A1
20060035258 Tadakamalla et al. Feb 2006 A1
20060040297 Leamon et al. Feb 2006 A1
20060041385 Bauer Feb 2006 A1
20060057634 Rye Mar 2006 A1
20060073506 Christians et al. Apr 2006 A1
20060211030 Brenner Sep 2006 A1
20060263709 Matsumura et al. Nov 2006 A1
20060263789 Kincaid Nov 2006 A1
20060280352 Muschler et al. Dec 2006 A1
20060281092 Wille et al. Dec 2006 A1
20060286570 Rowlen et al. Dec 2006 A1
20070020640 Mccloskey et al. Jan 2007 A1
20070031829 Yasuno et al. Feb 2007 A1
20070042400 Choi et al. Feb 2007 A1
20070042419 Barany et al. Feb 2007 A1
20070065823 Dressman et al. Mar 2007 A1
20070065844 Golub et al. Mar 2007 A1
20070105090 Cassidy et al. May 2007 A1
20070117121 Hutchison et al. May 2007 A1
20070117134 Kou May 2007 A1
20070117177 Luo et al. May 2007 A1
20070133856 Dutta-Choudhury Jun 2007 A1
20070172873 Brenner et al. Jul 2007 A1
20070178478 Dhallan et al. Aug 2007 A1
20070202523 Becker et al. Aug 2007 A1
20070281317 Becker et al. Dec 2007 A1
20080038727 Spier Feb 2008 A1
20080070303 West et al. Mar 2008 A1
20080119736 Dentinger May 2008 A1
20080194414 Albert et al. Aug 2008 A1
20080261204 Lexow Oct 2008 A1
20080268508 Sowlay Oct 2008 A1
20080269068 Church et al. Oct 2008 A1
20080274458 Latham et al. Nov 2008 A1
20080299609 Kwon et al. Dec 2008 A1
20080318802 Brenner Dec 2008 A1
20090053669 Liu et al. Feb 2009 A1
20090061513 Andersson et al. Mar 2009 A1
20090105959 Braverman et al. Apr 2009 A1
20090131269 Martin et al. May 2009 A1
20090137407 Church et al. May 2009 A1
20090220385 Brown et al. Sep 2009 A1
20090226891 Nova et al. Sep 2009 A2
20090252414 Suzuki Oct 2009 A1
20090253586 Nelson et al. Oct 2009 A1
20090283676 Skoglund Nov 2009 A1
20090290151 Agrawal et al. Nov 2009 A1
20090298709 Ma Dec 2009 A1
20090311694 Gallagher et al. Dec 2009 A1
20100069250 White, III Mar 2010 A1
20100105049 Ehrich et al. Apr 2010 A1
20100105112 Holtze et al. Apr 2010 A1
20100105886 Woudenberg et al. Apr 2010 A1
20100120098 Grunenwald et al. May 2010 A1
20100120630 Huang et al. May 2010 A1
20100136544 Agresti et al. Jun 2010 A1
20100159533 Lipson et al. Jun 2010 A1
20100167354 Kurn Jul 2010 A1
20100184076 Lawton Jul 2010 A1
20100255471 Clarke Oct 2010 A1
20100267028 Pasche Oct 2010 A1
20100291666 Collier et al. Nov 2010 A1
20100300895 Nobile et al. Dec 2010 A1
20100323348 Hamady et al. Dec 2010 A1
20100330574 Whitman Dec 2010 A1
20110038507 Hager Feb 2011 A1
20110059436 Hardin et al. Mar 2011 A1
20110059556 Strey et al. Mar 2011 A1
20110070584 Wohlgemuth et al. Mar 2011 A1
20110072889 Albitar et al. Mar 2011 A1
20110160078 Fodor et al. Jun 2011 A1
20110201507 Rava et al. Aug 2011 A1
20110230358 Rava Sep 2011 A1
20110244455 Larson et al. Oct 2011 A1
20110245111 Chee Oct 2011 A1
20110263457 Krutzik et al. Oct 2011 A1
20110294689 Namsaraev Dec 2011 A1
20110312511 Winquist et al. Dec 2011 A1
20120004132 Zhang et al. Jan 2012 A1
20120010091 Linnarson Jan 2012 A1
20120014977 Furihata et al. Jan 2012 A1
20120034607 Rothberg et al. Feb 2012 A1
20120040843 Ducree et al. Feb 2012 A1
20120045844 Rothberg et al. Feb 2012 A1
20120058520 Hayashida Mar 2012 A1
20120058902 Livingston et al. Mar 2012 A1
20120065081 Chee Mar 2012 A1
20120071331 Casbon Mar 2012 A1
20120087862 Hood et al. Apr 2012 A1
20120142018 Jiang Jun 2012 A1
20120149603 Cooney et al. Jun 2012 A1
20120156675 Lueerssen et al. Jun 2012 A1
20120163681 Lohse Jun 2012 A1
20120165219 Van Der Zaag et al. Jun 2012 A1
20120173159 Davey et al. Jul 2012 A1
20120190020 Oliphant et al. Jul 2012 A1
20120202293 Martin et al. Aug 2012 A1
20120220022 Ehrlich et al. Aug 2012 A1
20120220494 Samuels Aug 2012 A1
20120231972 Golyshin et al. Sep 2012 A1
20120252012 Armougom et al. Oct 2012 A1
20120253689 Rogan Oct 2012 A1
20120316074 Saxonov Dec 2012 A1
20120322681 Kung et al. Dec 2012 A1
20130005585 Anderson et al. Jan 2013 A1
20130022977 Lapidus et al. Jan 2013 A1
20130045994 Shinozuka et al. Feb 2013 A1
20130116130 Fu et al. May 2013 A1
20130190206 Leonard Jul 2013 A1
20130203047 Casbon et al. Aug 2013 A1
20130210643 Casbon et al. Aug 2013 A1
20130210659 Watson et al. Aug 2013 A1
20130224743 Casbon et al. Aug 2013 A1
20130225418 Watson Aug 2013 A1
20130225623 Buxbaum et al. Aug 2013 A1
20130237458 Casbon et al. Sep 2013 A1
20130267424 Casbon et al. Oct 2013 A1
20130274117 Church Oct 2013 A1
20130323732 Anderson et al. Dec 2013 A1
20140004569 Lambowitz et al. Jan 2014 A1
20140057799 Johnson et al. Feb 2014 A1
20140066318 Frisen et al. Mar 2014 A1
20140147860 Kaduchak et al. May 2014 A1
20140155274 Xie et al. Jun 2014 A1
20140155295 Hindson et al. Jun 2014 A1
20140178438 Sahin et al. Jun 2014 A1
20140194324 Gormley et al. Jul 2014 A1
20140206079 Malinoski et al. Jul 2014 A1
20140206547 Wang Jul 2014 A1
20140216128 Neat Aug 2014 A1
20140227684 Hindson et al. Aug 2014 A1
20140227705 Vogelstein et al. Aug 2014 A1
20140228239 McCoy et al. Aug 2014 A1
20140228255 Hindson et al. Aug 2014 A1
20140235506 Hindson et al. Aug 2014 A1
20140243242 Nicol et al. Aug 2014 A1
20140244742 Yu et al. Aug 2014 A1
20140272952 May et al. Sep 2014 A1
20140274811 Arnold Sep 2014 A1
20140287963 Hindson et al. Sep 2014 A1
20140303005 Samuels et al. Oct 2014 A1
20140309945 Park et al. Oct 2014 A1
20140315211 Sugino et al. Oct 2014 A1
20140322716 Robins Oct 2014 A1
20140357500 Vigneault et al. Dec 2014 A1
20140378322 Hindson et al. Dec 2014 A1
20140378345 Hindson et al. Dec 2014 A1
20140378349 Hindson et al. Dec 2014 A1
20140378350 Hindson et al. Dec 2014 A1
20150005199 Hindson et al. Jan 2015 A1
20150005200 Hindson et al. Jan 2015 A1
20150017654 Gorfinkel et al. Jan 2015 A1
20150066385 Schnall-levin et al. Mar 2015 A1
20150072867 Soldatov et al. Mar 2015 A1
20150099661 Fodor et al. Apr 2015 A1
20150099673 Fodor et al. Apr 2015 A1
20150111256 Church et al. Apr 2015 A1
20150118680 Fodor et al. Apr 2015 A1
20150119255 Fodor et al. Apr 2015 A1
20150119256 Fodor et al. Apr 2015 A1
20150119257 Fodor et al. Apr 2015 A1
20150119258 Fodor et al. Apr 2015 A1
20150119290 Fodor et al. Apr 2015 A1
20150133319 Fu et al. May 2015 A1
20150141292 Fodor et al. May 2015 A1
20150152409 Seitz et al. Jun 2015 A1
20150203897 Robons et al. Jul 2015 A1
20150211050 Iafrate et al. Jul 2015 A1
20150218620 Behlke Aug 2015 A1
20150225777 Hindson et al. Aug 2015 A1
20150225778 Hindson et al. Aug 2015 A1
20150247182 Faham et al. Sep 2015 A1
20150259734 Asbury et al. Sep 2015 A1
20150275295 Wang et al. Oct 2015 A1
20150298091 Weitz Oct 2015 A1
20150299784 Fan Oct 2015 A1
20150307874 Jaitin Oct 2015 A1
20150329852 Nolan Nov 2015 A1
20150360193 Fan et al. Dec 2015 A1
20150376609 Hindson et al. Dec 2015 A1
20160017320 Wang et al. Jan 2016 A1
20160026758 Jabara et al. Jan 2016 A1
20160053253 Salathia et al. Feb 2016 A1
20160055632 Fu et al. Feb 2016 A1
20160060621 Agresti et al. Mar 2016 A1
20160060682 Pregibon et al. Mar 2016 A1
20160068889 Gole et al. Mar 2016 A1
20160122751 LaBaer May 2016 A1
20160122753 Mikkelsen et al. May 2016 A1
20160138091 Chee et al. May 2016 A1
20160145677 Chee et al. May 2016 A1
20160145683 Fan et al. May 2016 A1
20160201125 Samuels et al. Jul 2016 A1
20160208322 Anderson et al. Jul 2016 A1
20160222378 Fodor et al. Aug 2016 A1
20160232291 Kyriazopoulou-Panagiotopoulou et al. Aug 2016 A1
20160244742 Linnarsson et al. Aug 2016 A1
20160244828 Mason Aug 2016 A1
20160257993 Fu et al. Sep 2016 A1
20160258012 Fodor et al. Sep 2016 A2
20160265027 Sanches-Kuiper et al. Sep 2016 A1
20160265069 Fan et al. Sep 2016 A1
20160289669 Fan et al. Oct 2016 A1
20160289670 Samuels et al. Oct 2016 A1
20160289740 Fu et al. Oct 2016 A1
20160298180 Chee Oct 2016 A1
20160312276 Fu et al. Oct 2016 A1
20160320720 Fan et al. Nov 2016 A1
20160326584 Fodor et al. Nov 2016 A1
20160355879 Kamberov et al. Dec 2016 A1
20160376583 Fodor et al. Dec 2016 A1
20160376648 Fodor et al. Dec 2016 A1
20170044525 Kaper et al. Feb 2017 A1
20170073730 Betts et al. Mar 2017 A1
20170154421 Fu et al. Jun 2017 A1
20170192013 Agresti et al. Jul 2017 A1
20170260584 Zheng et al. Sep 2017 A1
20170275669 Weissleder et al. Sep 2017 A1
20170314067 Shum et al. Nov 2017 A1
20170337459 Fodor et al. Nov 2017 A1
20170342405 Fu et al. Nov 2017 A1
20170342465 Shum et al. Nov 2017 A1
20170342484 Shum et al. Nov 2017 A1
20170344866 Fan et al. Nov 2017 A1
20180002764 Fan et al. Jan 2018 A1
20180016634 Hindson et al. Jan 2018 A1
20180024139 Peikon et al. Jan 2018 A1
20180030522 Kamberov et al. Feb 2018 A1
20180037942 Fu et al. Feb 2018 A1
20180057873 Zhou et al. Mar 2018 A1
20180088112 Fan et al. Mar 2018 A1
20180094312 Hindson et al. Apr 2018 A1
20180094314 Hindson et al. Apr 2018 A1
20180094315 Hindson et al. Apr 2018 A1
20180105808 Mikkelsen et al. Apr 2018 A1
20180112266 Hindson et al. Apr 2018 A1
20180127743 Vigneault et al. May 2018 A1
20180142292 Hindson et al. May 2018 A1
20180163201 Larson Jun 2018 A1
20180179590 Belgrader et al. Jun 2018 A1
20180179591 Belgrader et al. Jun 2018 A1
20180201923 LaBaer Jul 2018 A1
20180201980 Chee et al. Jul 2018 A1
20180208975 Peterson et al. Jul 2018 A1
20180216174 Shum et al. Aug 2018 A1
20180230527 Fang et al. Aug 2018 A1
20180251825 Stoeckius et al. Sep 2018 A1
20180258482 Hindson et al. Sep 2018 A1
20180258500 Fan et al. Sep 2018 A1
20180267036 Fan et al. Sep 2018 A1
20180276332 Fan et al. Sep 2018 A1
20180282803 Belgrader et al. Oct 2018 A1
20180291470 Fan et al. Oct 2018 A1
20180002738 Wang et al. Nov 2018 A1
20180320241 Nolan et al. Nov 2018 A1
20180327835 Fodor et al. Nov 2018 A1
20180327836 Fodor et al. Nov 2018 A1
20180327866 Fan et al. Nov 2018 A1
20180327867 Fan et al. Nov 2018 A1
20180340170 Belhocine et al. Nov 2018 A1
20180346969 Chang et al. Dec 2018 A1
20180346970 Chang et al. Dec 2018 A1
20180371536 Fu et al. Dec 2018 A1
20190025304 Vigneault et al. Jan 2019 A1
20190032129 Hindson et al. Jan 2019 A1
20190040474 Fan et al. Feb 2019 A1
20190085412 Fan et al. Mar 2019 A1
20190095578 Shum et al. Mar 2019 A1
20190100798 Fodor et al. Apr 2019 A1
20190119726 Shum et al. Apr 2019 A1
20190136316 Hindson et al. May 2019 A1
20190136317 Hindson et al. May 2019 A1
20190136319 Hindson et al. May 2019 A1
20190161743 Church et al. May 2019 A1
20190177788 Hindson et al. Jun 2019 A1
20190177800 Boutet et al. Jun 2019 A1
20190203270 Amit et al. Jul 2019 A1
20190203291 Hindson et al. Jul 2019 A1
20190211395 Tsao et al. Jul 2019 A1
20190218276 Regev et al. Jul 2019 A1
20190218607 Love et al. Jul 2019 A1
20190221287 Tsujimoto Jul 2019 A1
20190221292 Tsujimoto Jul 2019 A1
20190256888 Weissleder et al. Aug 2019 A1
20190256907 Ryan et al. Aug 2019 A1
20190292592 Shum et al. Sep 2019 A1
20190338278 Shum et al. Nov 2019 A1
20190338353 Belgrader et al. Nov 2019 A1
20190338357 Fan et al. Nov 2019 A1
20200109437 Chang et al. Apr 2020 A1
20200124601 Fan et al. Apr 2020 A1
20200149037 Shum May 2020 A1
20210198754 Fan et al. Jul 2021 A1
20210214770 Prosen et al. Jul 2021 A1
20210214784 Prosen et al. Jul 2021 A1
20210222244 Martin et al. Jul 2021 A1
20210230582 Fu et al. Jul 2021 A1
20210230583 Lam et al. Jul 2021 A1
20210230666 Wu et al. Jul 2021 A1
20210246492 Song et al. Aug 2021 A1
20210263019 Martin et al. Aug 2021 A1
20210355484 Jensen et al. Nov 2021 A1
20210371909 Lazaruk Dec 2021 A1
20220010361 Song et al. Jan 2022 A1
20220010362 Campbell Jan 2022 A1
Foreign Referenced Citations (238)
Number Date Country
2474509 Feb 2003 CA
102008025656 Dec 2009 DE
0 799 897 Oct 1997 EP
1 473 080 Nov 2004 EP
1 647 600 Apr 2006 EP
1 845 160 Oct 2007 EP
2036989 Mar 2009 EP
1379693 May 2009 EP
2204456 Jul 2010 EP
2431465 Mar 2012 EP
2203749 Aug 2012 EP
2511708 Oct 2012 EP
2538220 Dec 2012 EP
2 623 613 Aug 2013 EP
1745155 Oct 2014 EP
2 805 769 Nov 2014 EP
2556171 Sep 2015 EP
2970958 Dec 2017 EP
3263715 Jan 2018 EP
3136103 Aug 2018 EP
3256606 Aug 2018 EP
2954102 Dec 2018 EP
3428290 Jan 2019 EP
2970957 Apr 2019 EP
3058092 May 2019 EP
3327123 Aug 2019 EP
2293238 Mar 1996 GB
H04108385 Apr 1992 JP
2001078768 Mar 2001 JP
2005233974 Sep 2005 JP
2007504831 Mar 2007 JP
2008256428 Oct 2008 JP
2013039275 Feb 2013 JP
WO1989001050 Feb 1989 WO
WO1996024061 Aug 1996 WO
WO 9710365 Mar 1997 WO
WO 9915702 Apr 1999 WO
WO 9928505 Jun 1999 WO
WO 0058516 Oct 2000 WO
WO2001048242 Jul 2001 WO
WO2001053539 Jul 2001 WO
WO2002018643 Mar 2002 WO
WO2002046472 Jun 2002 WO
WO 02056014 Jul 2002 WO
WO2002059355 Aug 2002 WO
WO2002070684 Sep 2002 WO
WO2002072772 Sep 2002 WO
WO2002079490 Oct 2002 WO
WO2002083922 Oct 2002 WO
WO2002101358 Dec 2002 WO
WO2003035829 May 2003 WO
WO 04017374 Feb 2004 WO
WO2004021986 Mar 2004 WO
WO2004033669 Apr 2004 WO
WO2004066185 Aug 2004 WO
WO2004081225 Sep 2004 WO
WO2005017206 Feb 2005 WO
WO2005021731 Mar 2005 WO
WO 05042759 May 2005 WO
WO 05071110 Aug 2005 WO
WO 05080604 Sep 2005 WO
WO 05111242 Nov 2005 WO
WO2005111243 Nov 2005 WO
WO2006026828 Mar 2006 WO
WO 06071776 Jul 2006 WO
WO 06102264 Sep 2006 WO
WO2006137932 Dec 2006 WO
WO 07087310 Aug 2007 WO
WO 07087312 Aug 2007 WO
WO2007147079 Dec 2007 WO
WO2008047428 Apr 2008 WO
WO2008051928 May 2008 WO
WO2008057163 May 2008 WO
WO 08096318 Aug 2008 WO
WO2008104380 Sep 2008 WO
WO2008147428 Dec 2008 WO
WO2008150432 Dec 2008 WO
WO2009048530 Apr 2009 WO
WO 09148560 Dec 2009 WO
WO 09152928 Dec 2009 WO
WO2010059820 May 2010 WO
WO 10117620 Oct 2010 WO
WO2010131645 Nov 2010 WO
WO2011091393 Jul 2011 WO
WO2011106738 Sep 2011 WO
WO 11123246 Oct 2011 WO
WO2011127099 Oct 2011 WO
WO 11143659 Nov 2011 WO
WO 11155833 Dec 2011 WO
WO 12038839 Mar 2012 WO
WO 12042374 Apr 2012 WO
WO 12047297 Apr 2012 WO
WO 12048341 Apr 2012 WO
WO2012041802 May 2012 WO
WO 12083225 Jun 2012 WO
WO2012099896 Jul 2012 WO
WO 12108864 Aug 2012 WO
WO2012103154 Aug 2012 WO
WO2012112804 Aug 2012 WO
WO 12129363 Sep 2012 WO
WO 12140224 Oct 2012 WO
WO 12142213 Oct 2012 WO
WO 12148477 Nov 2012 WO
WO 12149042 Nov 2012 WO
WO 12162267 Nov 2012 WO
WO2012148497 Nov 2012 WO
WO2012156744 Nov 2012 WO
WO2012177639 Dec 2012 WO
WO 13019075 Feb 2013 WO
WO2013070990 May 2013 WO
WO2013096802 Jun 2013 WO
WO 13117595 Aug 2013 WO
WO 13130674 Sep 2013 WO
WO2013148525 Oct 2013 WO
WO 13173394 Nov 2013 WO
WO 13176767 Nov 2013 WO
WO 13177206 Nov 2013 WO
WO 13188831 Dec 2013 WO
WO 13188872 Dec 2013 WO
WO 13191775 Dec 2013 WO
WO 14015084 Jan 2014 WO
WO 14015098 Jan 2014 WO
WO 14018460 Jan 2014 WO
WO2014018093 Jan 2014 WO
WO 14028537 Feb 2014 WO
WO2014031997 Feb 2014 WO
WO 14071361 May 2014 WO
WO2014065756 May 2014 WO
WO 14093676 Jun 2014 WO
WO 14108850 Jul 2014 WO
WO 14124336 Aug 2014 WO
WO 14124338 Aug 2014 WO
WO 14126937 Aug 2014 WO
WO2014124046 Aug 2014 WO
WO 14144495 Sep 2014 WO
WO2014145458 Sep 2014 WO
WO2014176575 Oct 2014 WO
WO 14201273 Dec 2014 WO
WO 14210353 Dec 2014 WO
WO2014200767 Dec 2014 WO
WO2014204939 Dec 2014 WO
WO2014210223 Dec 2014 WO
WO2014210225 Dec 2014 WO
WO 15002908 Jan 2015 WO
WO 15031691 Mar 2015 WO
WO 15035087 Mar 2015 WO
WO 15044428 Apr 2015 WO
WO 15047186 Apr 2015 WO
WO2015057985 Apr 2015 WO
WO2015061844 May 2015 WO
WO 15103339 Jul 2015 WO
WO 15117163 Aug 2015 WO
WO2015134787 Sep 2015 WO
WO2015160439 Oct 2015 WO
WO2015168161 Nov 2015 WO
WO2015179339 Nov 2015 WO
WO 15200869 Dec 2015 WO
WO2015200893 Dec 2015 WO
WO2016044227 Mar 2016 WO
WO2016049418 Mar 2016 WO
WO2016061517 Apr 2016 WO
WO2016100976 Jun 2016 WO
WO2016118915 Jul 2016 WO
WO2016130578 Aug 2016 WO
WO2016160965 Aug 2016 WO
WO2016138496 Sep 2016 WO
WO2016138500 Sep 2016 WO
WO2016145409 Sep 2016 WO
WO2016149418 Sep 2016 WO
WO2016160844 Oct 2016 WO
WO2016168825 Oct 2016 WO
WO2016172373 Oct 2016 WO
WO2016190795 Dec 2016 WO
WO2016191272 Dec 2016 WO
WO2017032808 Mar 2017 WO
WO2017040306 Mar 2017 WO
WO2017044574 Mar 2017 WO
WO2017053905 Mar 2017 WO
WO2017079593 May 2017 WO
WO2017087873 May 2017 WO
WO2017096239 Jun 2017 WO
WO2017097939 Jun 2017 WO
WO2017117358 Jul 2017 WO
WO2017125508 Jul 2017 WO
WO2017139690 Aug 2017 WO
WO2017164936 Sep 2017 WO
WO2017173328 Oct 2017 WO
WO2017205691 Nov 2017 WO
WO2018017949 Jan 2018 WO
WO2018020489 Feb 2018 WO
WO2018031631 Feb 2018 WO
WO2018058073 Mar 2018 WO
WO2018064640 Apr 2018 WO
WO2018075693 Apr 2018 WO
WO2018111872 Jun 2018 WO
WO2018115852 Jun 2018 WO
WO2018119447 Jun 2018 WO
WO2018132635 Jul 2018 WO
WO2018140966 Aug 2018 WO
WO2018144240 Aug 2018 WO
WO2018144813 Aug 2018 WO
WO2018174827 Sep 2018 WO
WO2018217862 Nov 2018 WO
WO2018218222 Nov 2018 WO
WO2018222548 Dec 2018 WO
WO2018226293 Dec 2018 WO
WO2019055852 Mar 2019 WO
WO2019076768 Apr 2019 WO
WO2019084046 May 2019 WO
WO2019099906 May 2019 WO
WO2019113457 Jun 2019 WO
WO2019113499 Jun 2019 WO
WO2019113506 Jun 2019 WO
WO2019113533 Jun 2019 WO
WO2019118355 Jun 2019 WO
WO2019126789 Jun 2019 WO
WO2019157529 Aug 2019 WO
WO2013137737 Sep 2019 WO
WO2019178164 Sep 2019 WO
WO2019213237 Nov 2019 WO
WO2019213294 Nov 2019 WO
WO2020028266 Feb 2020 WO
WO2020033164 Feb 2020 WO
WO2020037065 Feb 2020 WO
WO2020046833 Mar 2020 WO
WO2020072380 Apr 2020 WO
WO2020097315 May 2020 WO
WO2020123384 Jun 2020 WO
WO2020154247 Jul 2020 WO
WO2020167920 Aug 2020 WO
WO2020214642 Oct 2020 WO
WO2021146207 Jul 2021 WO
WO2021146219 Jul 2021 WO
WO2021146636 Jul 2021 WO
WO2021155057 Aug 2021 WO
WO2021155284 Aug 2021 WO
WO2021163374 Aug 2021 WO
WO2021247593 Dec 2021 WO
Non-Patent Literature Citations (685)
Entry
Kang et al. Qncotarget 2015; 6: 13742-13749 (Year: 2015).
Lundberg et al. Nature Methods 2013; 10: 999-1002 + Online Methods and Supplementary Information (Year: 2013).
Macosko et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell 2015; 161: 1202-1214 (Year: 2015).
Vestheim et al. Application of Blocking Oligonucleotides to Improve Signal-to-Noise Ratio in a PCR. Methods in Molecular Biology 2011; 687: 265-274 (Year: 2011).
GenBank Accession No. NM_000518.5 for Homo sapiens hemoglobin subunit beta (HBB), mRNA. Mar. 22, 2021 [online], [retrieved on Apr. 14, 2021], retrieved from the Internet: <URL: www.ncbi.nlm.nih.gov/nuccore/NM_000518.5?report=Genbank (Year: 2021).
Achim et al., May 2015, High-throughput spatial mapping of single-cell RNA-seq data to tissue of origin. Nature Biotechnology, 33(5):503-511.
Alkan et al., Oct. 2009, Personalized copy number and segmental duplication maps using next-generation sequencing. Nat Genet., 41 (10):1061-1067.
Anderson, Feb. 11, 2014, Study describes RNA sequencing applications for molecular indexing methods, genomeweb.com, 5 pp.
Ansorge, 2009, Next-generation DNA sequencing techniques. New Biotechnology, 25(4):195-203.
Atanur et al., Jun. 2010, The genome sequence of the spontaneously hypertensive rat: Analysis and functional significance. Genome Res., 20(6):791-803.
Audic et al., 1997, The Significance of Digital Gene Expression Profiles. Genome Research, 7:986-995.
Bendall et al., May 6, 2011, Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science, 332(6030):687-696.
Bionumbers, Aug. 21, 2010, Useful fundamental numbers in molecular biology, http://bionumbers.hms.harvard.edu/KeyNumbers/aspx, 4 pp.
Bioscribe, Feb. 5, 2015, Massively parallel sequencing technology for single-cell gene expression published (press release), 3 pp.
Blainey, May 2013, The future is now: single-cell genomics of bacteria and archaea, FEMS Microbiol Rev., 37(3):407-427.
Bogdanova et al., Jan. 2008, Normalization of full-length enriched cDNA, Molecular Biosystems, 4(3):205-212.
Bonaldo et al., Sep. 1996, Normalization and subtraction: two approaches to facilitate gene discovery. Genome Res., 6(9):791-806.
Braha et al., 2000, Simultaneous stochastic sensing of divalent metal ions. Nature Biotechnology, 18:1005-1007.
Bratke et al., Sep. 2005, Differential expression of human granzymes A, B, and K in natural killer cells and during CD8+ T cell differentiation in peripheral blood. Eur J Immunol., 35(9):2608-2616.
Brenner et al., 2000, Gene expression analysis by massively parallel signature sequencing (MPSS) on microbead arrays. Nature Biotechnology, 18:630-634.
Brenner et al., Feb. 15, 2000, In vitro cloning of complex mixtures of DNA on microbeads: physical separation of differentially expressed cDNAs. Proc Natl Acad Sci, 97(4):1665-1670.
Brisco et al., Jun. 25, 2012, Quantification of RNA integrity and its use for measurement of transcript number, Nucleic Acids Research, 40(18):e144.
Brodin et al., 2015, Challenges with using primer IDs to improve accuracy of next generation sequencing, 19(3):1-12.
Butkus, Feb. 6, 2014, Cellular research set to launch first gene expression platform using ‘molecular indexing’ technology, genomeweb.com, 5 pp.
Cai, Mar. 2013, Turning single cells in microarrays by super-resolution bar-coding, Brief Funct Genomics, 12(2):75-80.
Carr et al., Dec. 15, 2009, Inferring relative proportions of DNA variants from sequencing electropherograms. Bioinformatics, 25(24):3244-3250.
Casbon et al., Jul. 2011, A method for counting PCR template molecules with application to next-generation sequencing. Nucleic Acids Res., 39(12):e81.
Castellarnau et al., Jan. 2015, Stochastic particle barcoding for single-cell tracking and multiparametric analysis, Smail, 11(4):489-498.
Castle et al., Apr. 16, 2010, DNA copy number, including telomeres and mitochondria, assayed using next-generation sequencing. BMC Genomics, 11:244. doi: 10.1186/1471-2164-11-244.
Chamberlain et al., Dec. 9, 1988, Deletion screening of the Duchenne muscular dystrophy locus via multiplex DNA amplification. Nucleic Acids Res., 16(23):11141-11156.
Chang et al., Aug. 2002, Detection of allelic imbalance in ascitic supernatant by digital single nucleotide polymorphism analysis. Clin Cancer Res., 8(8):2580-2585.
Chee et al., 1996, Accessing genetic information with high-density DNA arrays, Science, 274:610-614.
Chee, 1991, Enzymatic multiplex DNA sequencing. Nucleic Acids Research, 19(12): 3301-3305.
Chen et al., Apr. 9, 2015, Spatially resolved, highly multiplexed RNA profiling in single cells. Science Express, pp. 1-21.
Church et al., 1988, Multiplex DNA sequencing. Science, 240:185-188.
Costello et al., Apr. 1, 2013, Discovery and characterization of artefactual mutations in deep coverage targeted capture sequencing data due to oxidative DNA damage during sample preparation. Nucleic Acids Res, 41(6):e67.
Cox, May 2001, Bar coding objects with DNA. Analyst, 126(5):545-547.
Craig et al., Oct. 2008, Identification of genetic variants using bar-coded multiplexed sequencing. Nat Methods, 5(10):887-893.
Cusanovich et al., May 7, 2014, Multiplex single-cell profiling of chromatin accessibility by combinatorial cellular indexing. Science Express, pp. 1-9.
Daines et al., Aug. 2009, High-throughput multiplex sequencing to discover copy number variants in Drosophila. Genetics, 182(4):935-941.
Dalerba et al., 2011, Single-cell dissection of transcriptional heterogeneity in human colon tumors, Nat Biotechnol., 29(12):1120-1127 and Supplementary Material.
D'Antoni et al., May 1, 2006, Rapid quantitative analysis using a single molecule counting approach. Anal Biochem. 352(1):97-109.
Daser et al., 2006, Interrogation of genomes by molecular cooy-number counting (MCC). Nature Methods, 3(6):447-453.
De Saizieu et al., 1998, Bacterial transcript imaging by hybridization of total RNA to oligonucleotide arrays. Nature Biotechnology, 16:45-48.
Di Carlo et al., Dec. 1, 2008, Dynamic single-cell analysis for quantitative biology, Analytical Chemistry, 78(23):7918-7925.
Dirks et al., Oct. 26, 2004, Triggered amplification by hybridization chain reaction., Proc Natl Acad Sci U S A, 101(43), 15275-15278.
Fan et al., 2000, Parallel Genotyping of Human SNPs Using Generic High-density Oligonucleotide Tag Arrays. Genome Research, 10:853-860.
Fan et al., 2009, Microfluidic digital PCR enables rapid prenatal diagnosis of fetal aneuploidy. Am Obstet Gynecol. 200:543.e1-543.e7.
Fan et al., Feb. 6, 2015, Combinatorial labeling of single cells for gene expression cytometry. Science, 347(6222):1258367-8.
Fan et al., Jul. 19, 2012, Non-invasive prenatal measurement of the fetal genome. Nature, 487(7407):320-324.
Fan, Nov. 2010, Molecular counting: from noninvasive prenatal diagnostics to whole-genome haplotyping, doctoral dissertation, Stanford University, 185 pp.
Feldhaus et al., Jan. 15, 2000, Oligonucleotide-conjugated beads for transdominant genetic experiments, Nucleic Acids Res., 28(2):534-543.
Fox-Walsh et al., Oct. 2011, A multiplex RNA-seq strategy to profile poly(A+) RNA: application to analysis of transcription response and 3′ end formation., Genomics, 98(4),266-271.
Fu et al., Mar. 18, 2014, Digital encoding of cellular mRNAs enabling precise and absolute gene expression measurement by single-molecule counting. Anal Chem., 86(6):2867-2870.
Fu et al., May 31, 2011, Counting individual DNA molecules by the stochastic attachment of diverse labels. Proc Natl Acad Sci, 108(22):9026-9031.
Gerry et al., 1999, Universal DNA microarray method for multiplex detection of low abundance point mutations. Journal of Molecular Biology, 292(2): 251-262.
Gillespie, 1977, Exact stochastic simulation of coupled chemical reactions. The Journal of Physical Chemistry, 81(25):2340-2361.
Gong et al., 2010, Massively parallel detection of gene expression in single cells using subnanolitre wells, Lab Chip, 10:2334-2337.
Grant et al., Nov. 15, 2002, SNP genotyping on a genome-wide amplified DOP-PCR template. Nucleic Acids Res, 30(22):e125.
Gu et al., 2016, Depletion of abundant sequences by hybridization (DSH): using Cas9 to remove unwanted high-abundance species in sequencing libraries and molecular counting applications, Genome Biology, 17:41, 13 pp.
Gunderson et al., May 2004, Decoding randomly ordered DNA arrays. Genome Res. 14(5):870-877.
Gundry et al., Jan. 3, 2012, Direct mutation analysis by high-throughput sequencing: from germline to low-abundant, somatic variants. Mutat Res. 729(1-2):1-15.
Gundry et al., Mar. 2012, Direct, genome-wide assessment of DNA mutations in single cells. Nucleic Acids Res., 40(5):2032-40.
Hacia et al., 1999, Determination of ancestral alleles for human single-nucleotide polymorphisms using high-density oligonucleotide arrays. Nature Genetics, 22:164-167.
Haff, 1994, Improved quantitative PCR using nested primers, PCR Methods and Applications, 3:332-337.
Hamady et al., Mar. 2008, Error-correcting barcoded primers for pyrosequencing hundreds of samples in multiplex. Nat Methods, 5(3):235-237.
Harrington et al., 2009, Cross-sectional characterization of HIV-1 env compartmentalization in cerebrospinal fluid over the full disease course, AIDS, 23(8) 907-915.
Hashimshony et al., Sep. 27, 2012, CEL-Seq: single-cell RNA-Seq by multiplexed linear amplification Cell Rep. 2(3):666-673.
Hensel et al., Jul. 21, 1995, Simultaneous identification of bacterial virulence genes by negative selection. Science. 269(5222):400-403.
Hiatt et al., Feb. 2010, Parallel, tag-directed assembly of locally derived short sequence reads. Nat Methods, 7(2):119-122.
Hiatt et al., May 2013, Single molecule molecular inversion probes for targeted, high-accuracy detection of low-frequency variation. Genome Res., 23(5):843-854.
Hollas et al., 2003, A stochastic approach to count RNA molecules using DNA sequencing methods. Lecture Notes in Computer Science, 2812:55-62.
Hug et al., 2003, Measure of the number of molecular of a single mRNA species in a complex mRNA preparation, Journal of Theoretical Biology, 221:615-624.
Ingolia et al., Apr. 10, 2009, Genome-wide analysis in vivo of translation with nucleotide resolution using ribosome profiling. Science, 324(5924):218-223.
Islam et al., 2011, Characterization of the single-cell transcriptional landscape by highly multiplex RNA-seq. Genome Research, 21:1160-1167.
Islam et al., 2014, Quantitative single-cell RNA-seq with unique molecular identifiers, Nature Methods, 11(2):163-168.
Jabara et al., Dec. 3, 2011, Accurate sampling and deep sequencing of the HIV-1 protease gene using a Primer ID, PNAS, 108(50):20166-20171.
Jabara, Apr. 23, 2010, Capturing the cloud: High throughput sequencing of multiple individual genomes from a retroviral population. Biology Lunch Bunch Series, Training Initiatives in Biomedical & Biological Sciences of the University of North Carolina at Chapel Hill.
Junker et al., May 21, 2015, Single-cell transcriptomics enters the age of mass production, Molecular Cell, 58:563-564.
Kanagawa, 2003, Bias and artifacts in multitemplate polymerase chain reactions (PCR), Journal of Bioscience and Bioengineering, 96(4):317-323.
Kebschull et al., Jul. 17, 2015, Sources of PCR-induced distortions in high-throughput sequencing data sets, Nucleic Acids Research, 15 pp.
Keys et al., Jun. 2015, Primer ID informs next-gene ration sequencing platforms and reveals preexisting drug resistance mutations in the HIV-1 reverse transcriptase coding domain, AIDS Research and Human Retroviruses, 31(6):658-668.
Kim et al., Jun. 8, 2007, Polony multiplex analysis of gene expression (PMAGE) in mouse hypertrophic cardiomyopathy, Science, 316(5830):1481-1484.
Kinde et al., Jun. 7, 2011, Detection and quantification of rare mutations with massively parallel sequencing, Proc. Natl Acad Sci, 108(23):9530-0535.
Kivioja et al., Jan. 2012, Counting absolute numbers of molecules using unique molecular identifiers. Nature Methods, 9(1):72-76.
Klein et al., May 21, 2015, Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells, Cell, 161:1187-1201.
Koboldt et al., Sep. 1, 2009, VarScan: variant detection in massively parallel sequencing of individual and pooled samples. Bioinformatics. 25(17):2283-2285.
Kolodziejczyk et al., May 21, 2015, The technology and biology of single-cell RNA sequencing, Molecular Cell, 58:610-620.
Konig et al., Jul. 2010, iCLIP reveals the function of hnRNAP particles in splicing at individual nucleotide resolution, Nature Structural & Molecular Biology, 17(7):909-916.
Kotake et al., 1996, A simple nested RT-PCR method for quantitation of the relative amounts of multiple cytokine mRNAs in small tissue samples, Journal of Immunological Methods, 199:193-203.
Kurimoto et al., Mar. 17, 2006, An improved single-cell cDNA amplification method for efficient high-density oligonucleotide microarray analysis, Nucleic Acids Res., 34(5):e42.
Lamble et al., Nov. 20, 2013, Improved workflows for high throughput library preparation using the transposome-based nextera system, BMC Biotechnology, 13(1):104.
Larson et al., Nov. 2009, A single molecule view of gene expression. Trends Cell Biol. 19(11):630-637.
Leamon et al., Nov. 2003, A massively parallel PicoTiterPlate based platform for discrete picoliter-scale polymerase chain reactions, Electrophoresis, 24(21):3769-3777.
Lee et al., 2010, Large-scale arrays of picolitre chambers for single-cell analysis of large cell populations, Lab Chip, 10:2952-2958.
Lee et al., Mar. 21, 2014, Highly multiplexed subcellular RNA sequencing in situ. Science. 343(6177):1360-1363.
Liu et al., Single-cell transcriptome sequencing: recent advances and remaining challenges, F1000Research 2016, 5(F1000 Faculty Rev):182, 9 pp.
Lizardi et al., Jul. 1998, Mutation detection and single-moiecuie counting using isothermal rolling-circle amplification. Nat Genet. 19(3):225-32.
Lockhart et al., 1996, Expression monitoring by hybridization to high-density oligonucleotide arrays. Nature Biotechnology, 14:1675-1680.
Lovatt et al., Feb. 2014, Transcriptome in vivo analysis (TIVA) of spatially defined single cells in live tissue. Nat Methods. 11(2):190-196.
Lucito et al., 1996, Representational Oligonucleotide Microarray Analysis: A High-Resolution Method to Detect Genome Copy Number Variation. Genome Research, 13: 2291-2305.
Maamar et al., 2007, Noise in Gene Expression Determines Cell rate in Bacillus subtilis. Science, 317:526-529.
Macaulay et al., 2015, G&T-seq: parallel sequencing of single-cell genomes and transcriptomes. Nature Methods, pp. 1-7.
Macosko et al., 2015, Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets, Cell 161:1202-1214 (and supplemental information).
Makrigiorgos et al., Sep. 2002, A PCR-Based amplification method retaining quantities difference between two complex genomes. Nature Biotech, 20(9):936-939.
Marcus et al., 2006, Microfluidic single-cell mRNA isolation and analysis, Ana. Chem. 78:3084-3089.
Margulies et al., Sep. 15, 2005 Genome sequencing in microfabricated high-density picolitre reactors, Nature, 437:376-380.
Martinez et al., Jul. 2012, A microfluidic approach to encapsulate living cells in uniform alginate hydrogel microparticles, Macromol. Biosci, 12(7):946-951.
McCloskey et al., Dec. 2007, Encoding PCR products with batch-stamps and barcodes. Biochem Genet. 45(11-12):761-767.
Medvedev et al., Nov. 2010, Detecting copy number variation with mated short reads. Genome Res. 20(11):1613-1622.
Mei et al., Mar. 22, 2010, Identification of recurrent regions of Copy-Number Variants across multiple individuals. BMC Bioinformatics. 11:147.
Merriam-Webster, definition of associate,: http://www.merriam-webster.com/dictionary/associate, accessed Apr. 5, 2016.
Miller et al., 2006, Directed evolution by in vitro compartmentalization, Nature Methods, 3:561-570.
Miner et al., 2004, Molecular barcodes detect redundancy and contamination in hairpin-bisulfite PCR, Nucleic Acids Research, 32(17):e135.
Mortazavi et al., 2008, Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat. Methods. 5:621-628.
Nadai et al., 2008, Protocol for nearly full-length sequencing of HIV-1 RNA from plasma, PLoS One, 3(1):e1420.
Nagai et al., 2001, Development of a microchamber array for picoleter PCR, Anal. Chem., 73:1043-1047.
Navin et al., 2015, The first five years of single-cell cancer genomics and beyond, Genome Research, 25(10):1499-1507.
Newell et al., Jan. 27, 2012, Cytometry by time-of-flight shows combinatorial cytokine expression and virus-specific cell niches within a continuum of CD8+ T cell phenotypes. Immunity. 36(1):142-152.
Ogino et al., Nov. 2002, Quantification of PCR bias caused by a single nucleotide polymorphism in SMN gene dosage analysis. J Mol Diagn. 4(4):185-190.
Parameswaran et al., 2007, A pyrosequencing-tailored nucleotide barcode design unveils opportunities for large-scale sample multiplexing. Nucleic Acids Res. 35(19):e130.
Park et al., May 2010, Discovery of common Asian copy number variants using integrated high-resolution array CGH and massively parallel DNA sequencing. Nat Genet. 42(5):400-405.
Patanjali et al., Mar. 1991, Construction of a uniform-abundance (normalized) CNDA library, Proceedings of the National Academy of Sciences, 88(5):1943-1947.
Peng et al., Mar. 11, 2016, Reducing amplification artifacts in high multiplex amplicon sequencing by using molecular barcodes, BMC Genomics, retrieved from the internet: url:http://bmcgenomics.biomedcentral.com/aricles/0.1186/s12864-015-1806-8, 14 pp.
Pfaffl et al., Mar. 2004, Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper—Excel-based tool using pair-wise correlations, Biotechnology Letters, 26(6):505-515.
Picelli et al., Jul. 30, 2014, Tn5 transposase and tagmentation procedures for massively scaled sequencing projects, Genome Research 24(12):2033-2040.
Pihlak et al., 2008, Rapid genome sequencing with short universal tiling probes. Nature Biotechnology, 26:676-684.
Pinkel et al., 2005, Comparative Genomic Hybridization. Annual Review of Genomics and Human Genetics, 6:331-354.
Pleasance et al., Jan. 14, 2010, A small-cell lung cancer genome with complex signatures of tobacco exposure. Nature. 463(7278):184-190.
Plessy et al., Feb. 2013, Population transcriptomics with single-cell resolution: a new field made possible by microfluidics: a technology for high throughput transcript counting and data-driven definition of cell types, Bioessays, 35(2):131-140.
Qiu et al., Oct. 2003, DNA sequence-based “barcodes” for tracking the origins of expressed sequence tags from a maize cDNA library constructed using multiple mRNA sources. Plant Physiol. 133(2):475-481.
Rajeevan et al., Oct. 2003, Global amplification of sense RNA: a novel method to replicate and archive mRNA for gene expression analysis, Genomics, 82(4):491-497.
Roche Diagnostics GmbH, 2006, Genome Sequencer 20 System: First to the Finish (product brochure), 40 pp.
Sasagawa et al., 2013, Quartz-Seq: a highly reproducible and sensitive single-cell RNA sequencing method, reveals non-genetic gene-expression heterogeneity. Genome Biology, 14:R31.
Sasuga et al., Dec. 2008, Single-cell chemical lysis method for analyses of intracellular molecules using an array of picoliter-scale microwells, Anal Chem, 80(23):9141-9149.
Satija et al., May 2015, Spatial reconstruction of single-cell gene expression data. Nature Biotechnology, 33(5):495-508.
Schmitt et al., Sep. 4, 2012, Detection of ultra-rare mutations by next-generation sequencing. Proc Natl Acad Sci U S A. 109(36):14508-14513.
Sebat et al., 2004, Large-Scale Copy Number Polymorphism in the Human Genome. Science, 305:525-528.
Shalek et al., Jun. 13, 2013, Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature. 498(7453):236-240.
Shiroguchi et al., Jan. 24, 2012, Digital RNA sequencing minimizes sequence-dependent bias and amplification noise with optimized single-molecule barcodes. Proc Natl Acad Sci USA. 109(4):1347-1352.
Shoemaker et al., 1996, Quantitative phenotypic analysis of yeast deletion mutants using a highly parallel molecular bar-coding strategy. Nature Genetics, 14:450-456.
Simpson et al., Feb. 15, 2010, Copy number variant detection in inbred strains from short read sequence data. Bioinformatics. 26(4):565-567.
Smith et al., 2010, Highly-multiplexed barcode sequencing: an efficient method for parallel analysis of pooled samples. Nucleic Acids Research. 38(13):e142.
Soumillon et al., Mar. 5, 2014, Characterization of directed differentiation by high-throughput single-cell RNA-Seq, bioRxiv preprint, http://biorxiv.org/content/early/2014/03/05/003236.full.pdf, 13 pp.
Speicher et al., Oct. 2005, The new cytogenetics: blurring the boundaries with molecular biology, Nature Reviews Genetics, 6(10):782-792.
Stratagene 1998 Catalog, Gene Characterization Kits, p. 39.
Takahashi et al., Mar. 2006, Novel technique of quantitative nested real-time PCR assay for Mycobacterium tuberculosis DNA, Journal of Clinical Microbiology, 44(3):1029-1039.
Tan et al., Apr. 2013, Genome-wide comparison of DNA hydroxymethylation in mouse embryonic stem cells and neural progenitor cells by a new comparative hMeDIP-seq method. Nucleic Acids Res. 41(7):e84.
Taudien et al., Apr. 19, 2010, Haplotyping and copy number estimation of the highly polymorphic human beta-defensin locus on 8p23 by 454 amplicon sequencing. BMC Genomics. 11:252.
The Tibbs Times, UNC bioscience newsletter, Apr. 2010, 17 pp.
Tomaz et al., Aug. 2010, Differential methylation as a cause of allele dropout at the imprinted GNAS locus. Genet Test Mol Biomarkers. 14(4):455-460.
Treutlein et al., May 15, 2014, Reconstructing lineage hierarchies of the distal lung epithelium using single-cell RNA-seq. Nature. 509(7500):371-375.
Vandesompele et al., Jun. 18, 2002, Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes, Genome Biology, 3(7).
Velculescu et al., 1995, Serial Analysis of Gene Expression. Science, 270:484-487.
Velculescu et al., 1997, Characterization of the Yeast Transcriptome. Cell, 88:243-251.
Vogelstein et al., 1999, Digital PCR. Proc. Natl. Acad. Sci., 96(16):9236-9241.
Walker et al., Jan. 1, 1992, Isothermal in vitro amplification of DNA by a restriction enzyme/DNA polymerase system. Proc Natl Acad Sci U S A., 89(1):392-396.
Walsh et al., Jul. 13, 2010, Detection of inherited mutations for breast and ovarian cancer using genomic capture and massively parallel sequencing. Proc Natl Acad Sci USA. 107(28):12629-12633.
Wang et al., 2009, RNA-Seq: a revolutionary tool for transcriptomics. Nature Reviews Genetics, 10:57-63.
Wang et al., May 21, 2015, Advances and applications of single-cell sequencing technologies, Molecular Cell, 58(4):598-609.
Wang et al., Oct. 2010, iCLIP predicts the dual splicing effects of TIA-RNA interactions, PLoS Biol, 8(10):e1000530.
Warren et al., Nov. 21, 2006, Transcription factor profiling in individual hematopoietic progenitors by digital RT-PCR, PNAS, 103(47):17807-17812.
Weber et al., Sep. 15, 2003, A real-time polymerase chain reaction assay for quantification of allele ratios and correction of amplification bias. Anal Biochem. 320(2):252-258.
Weiner et al., Apr. 2008, Kits and their unique role in molecular biology: a brief retrospective, BioTechniques, 44:701-704.
White et al., Aug. 23, 2011, High-throughput microfluidic single-cell RT-qPCR, PNAS, 108(34):13999-14004.
Wittes et al., 1999, Searching for Evidence of Altered Gene Expression: a Comment on Statistical Analysis of Microarray Data. Journal of the National Cancer Institute, 91(5):400-401.
Wodicka et al., 1997, Genome-wide expression monitoring in Saccharomyces cerevisiae. Nature Biotechnology, 15:1359-1367.
Wojdacz et al., May 16, 2009, Primer design versus PCR bias in methylation independent PCR amplifications. Epigenetics. 4(4):231-234.
Wood et al., Aug. 2010, Using next-generation sequencing for high resolution multiplex analysis of copy number variation from nanogram quantities of DNA from formalin-fixed paraffin-embedded specimens. Nucleic Acids Res. 38(14):e151.
Wu et al., Jan. 2014, Quantitative assessment of single-cell RNA-sequencing methods. Nat Methods. 11(1):41-46.
Yandell et al., Sep. 2011, A probabilistic disease-gene finder for personal genomes. Genome Res. 21(9):1529-1542.
Ye et al., 2001, Fluorescent microsphere-based readout technology for multiplexed human single nucleotide polymorphism analysis and bacterial identification. Human Mutation, 17(4):305-316.
Yoon et al., Sep. 2009, Sensitive and accurate detection of copy number variants using read depth of coverage. Genome Res. 19(9):1586-1592.
Zhang et al., Jun. 19, 2012, DNA-based hybridization chain reaction for amplified bioelectronic signal and ultrasensitive detection of proteins. Anal Chem., 84(12),5392-5399.
Zhang et al., Mar. 20, 2011, The impact of next-generation sequencing on genomics. J Genet Genomics. 38(3):95-109.
Zhao et al., 2005, Homozygous Deletions and Chromosome Amplifications in Human Lung Carcinomas Revealed by Single Nucleotide Polymorphism Array Analysis. Cancer Research, 65:5561-5570.
Zheng et al., Feb. 2016, Haplotyping germline and cancer genomes with high-throughput linked-read sequencing, Nature Biotechnology, 34(3):303-311.
Zhou et al., 2001, Counting alleles reveals a connection between chromosome 18q loss and vascular invasion. Nature Biotechnology, 19:78-81.
International Search Report and Written Opinion dated May 3, 2016 in PCT/US16/018354.
Office action dated Oct. 3, 2013 for U.S. Appl. No. 12/969,581.
Response with allowed claims dated Mar. 4, 2014 for U.S. Appl. No. 12/969,581.
Notice of allowance dated Mar. 21, 2014 for U.S. Appl. No. 12/969,581.
Notice of allowance dated Jun. 19, 2014 for U.S. Appl. No. 12/969,581.
Notice of allowance dated Aug. 22, 2014 for U.S. Appl. No. 12/969,581.
Office action dated Dec. 3, 2015 for U.S. Appl. No. 14/281,706.
Office action dated Jul. 20, 2016 for U.S. Appl. No. 14/281,706.
Office Action dated Oct. 11, 2016 in U.S. Appl. No. 15/224,460.
Office Action dated May 8, 2017 in U.S. Appl. No. 15/224,460.
Office Action dated May 7, 2015 for U.S. Appl. No. 13/327,526.
Notice of allowance dated Jan. 21, 2016 for U.S. Appl. No. 13/327,526.
Office Action dated Jul. 28, 2017 in U.S. Appl. No. 14/975,441.
Office action dated Feb. 18, 2015 for U.S. Appl. No. 14/540,007.
Office action dated Sep. 24, 2015 for U.S. Appl. No. 14/540,007.
Notice of allowance dated Dec. 15, 2015 for U.S. Appl. No. 14/540,007.
Office action dated Mar. 19, 2015 for U.S. Appl. No. 14/540,018.
Office action dated Oct. 6, 2015 for U.S. Appl. No. 14/540,018.
Notice of allowance dated Dec. 21, 2015 for U.S. Appl. No. 14/540,018.
Office Action dated Feb. 26, 2015 for U.S. Appl. No. 14/540,029.
Office action dated Sep. 1, 2015 for U.S. Appl. No. 14/540,029.
International Search Report and Written Opinion dated Jun. 6, 2012 in PCT/US11/065291.
Office Action dated Feb. 17, 2017 in Canadian patent application No. 2,865,575.
Office Action dated Jun. 6, 2016 in Chinese patent application No. 201380022187.9.
Office Action dated Dec. 27, 2016 in Chinese patent application No. 201380022187.9.
Office Action dated Jul. 14, 2017 in Chinese patent application No. 201380022187.9.
European search report and search opinion dated Jul. 17, 2015 for European patent application No. 13755319.4.
Examination report dated Jul. 12, 2016 in European patent application No. 13755319.4.
Search and Examination Report dated Aug. 6, 2014 for GB patent application No. 1408829.8.
Search and Examination Report dated Jan. 27, 2016 in GB patent application No. 1408829.8.
Examination Report dated Jun. 8, 2016 in GB patent application No. 1408829.8.
Official Action dated Dec. 28, 2016 in Japanese patent application No. 2014-558975.
Restriction Requirement dated Mar. 15, 2016 in U.S. Appl. No. 14/381,488.
Office Action dated May 10, 2016 in U.S, U.S. Appl. No. 14/381,488.
Office Action dated Aug. 12, 2016 in U.S. Appl. No. 14/381,488.
Office Action dated Feb. 13, 2017 in U.S. Appl. No. 14/381,488.
Office Action dated Jun. 7, 2017 in U.S. Appl. No. 14/381,488.
Search Report and Written Opinion dated Mar. 1, 2016 in Singapore patent application No. 11201405274W.
Written Opinion dated May 26, 2017 in Singapore patent application No. 11201405274W.
International Search Report and Written Opinion dated Sep. 6, 2013 in PCT/US13/028103.
Extended European Search Report dated Dec. 15, 2015 in European patent application No. 13754428.4.
International search report and written opinion dated Aug. 16, 2013 for PCT/US2013/027891.
Office Action dated Apr. 11, 2016 in U.S. Appl. No. 14/472,363.
Restriction Requirement dated Mar. 17, 2016 in U.S. Appl. No. 14/472,363.
Office action dated Dec. 31, 2015 for U.S. Appl. No. 14/800,526.
Office action dated Apr. 11, 2016 for U.S. Appl. No. 14/800,526.
Office action dated Aug. 17, 2016 for U.S. Appl. No. 14/800,526.
Office Action dated Oct. 25, 2016 in U.S. Appl. No. 14/872,337.
Office action dated Sep. 26, 2016 in U.S. Appl. No. 15/167,807.
Examination Report dated Apr. 10, 2017 in European patent application No. 14761937.3.
Search and Examination Report dated Aug. 26, 2015 in GB patent application No. 1511591.8.
Examination Report dated Feb. 19, 2016 in Great Britain patent application No. GB1511591.8.
Examination Report dated Jun. 15, 2016 in Great Britain patent application No. GB1511591.8.
Combined Search and Examination Report dated Feb. 21, 2017 in GB patent application No. 1609740.4.
International Search Report and Written Opinion dated Feb. 3, 2015 in PCT/US/14/053301.
Third Party Observation dated Jun. 14, 2018 in Japanese patent application No. 2016-537867.
Official Action dated Jul. 30, 2018 in Japanese patent application No. 2016-537867.
Office Action dated May 13, 2016 in U.S. Appl. No. 14/508,911.
Office Action dated Mar. 24, 2017 in U.S. Appl. No. 15/409,355.
International search report and written opinion dated Dec. 19, 2014 for PCT Application No. US2014/059542.
International Search Report and Written Opinion dated Jun. 20, 2016 in PCT/US16/14612.
Office Action dated Jan. 19, 2017 in U.S. Appl. No. 15/055,445.
International Search Report and Written Opinion dated Jun. 17, 2016 in PCT/US16/019962.
Written Opinion dated Jul. 5, 2016 in PCT/US16/019962.
Written Opinion dated Sep. 27, 2016 in PCT/US16/019962.
Invitation to Pay Additional Search Fees dated Jun. 2, 2016 in PCT/US16/019971.
International Search Report and Written Opinion dated Aug. 9, 2016 in PCT/US16/019971.
International Search Report and Written Opinion dated Jun. 9, 2016 in PCT/US16/022712.
International Search Report and Written Opinion dated Dec. 5, 2016 in PCT/US16/024783.
International Search Report and Written Opinion dated Sep. 28, 2016 in PCT/US16/028694.
International Search Report and Written Opinion dated Sep. 27, 2016 in PCT/US16/034473.
International Search Report and Written Opinion dated Jan. 31, 2017 in PCT/US16/050694.
International Search Report and Written Opinion dated Aug. 7, 2017 in PCT/US2017/034576.
Office Action dated Apr. 6, 2018 in U.S. Appl. No. 15/603,239.
International Search Report and Written Opinion dated Mar. 28, 2018 in patent application No. PCT/US2018/014385.
International search report and written opinion dated May 7, 2012 for PCT/IB2011/003160.
Notice of opposition dated Jul. 22, 2015 for European patent application No. 11810645.9.
Notice of opposition dated Jul. 9, 2015 for European patent application No. 11810645.9.
10X Genomics, Inc., 2019, User Guide: Visium Spatial Gene Expression Reagent Kits, www.10xGenomics.com, 76 pp.
2018 Top 10 Innovations, The Scientist Magazine® (2018). Available at: https://www.thescientist.com/features/2018-top-10-innovations-65140, 16 pp.
Advisory Action dated Dec. 2, 2019 in U.S. Appl. No. 15/055,407.
Advisory Action dated Nov. 29, 2019 in U.S. Appl. No. 15/084,307.
Agasti et al., “Photocleavable DNA barcode-antibody conjugates allow sensitive and multiplexed protein analysis in single cell,” J Am Chem Soc. 2012, 134(45), 18499-18502.
Alexandra M. Ewing of Richards, Layton and Finger, P.A., Entry of Appearance dated Jan. 18, 2019 in the USDC District of Delaware, C.A. No. 18-1800-RGA, 1 pp.
Applied Biosystems, Apr. 2008, SOLiDTM System Barcoding, Application Note, 4 pp.
Argrawal et al., “Counting Single Native Biomolecules and Intact Viruses with Color-Coded Nanoparticles,” Analytical Chemistry 2006, 78, 1061-1070.
Arslan et al., “An efficient algorithm for the stochastic simulation of the hybridization of DNA to microarrays,” BMC Bioinformatics 2009, 10(411), 1-17.
Baek et al., “Development of Hydrogel TentaGel Shell-Core Beads for Ultra-high Throughput Solution Phase Screening of Encoded OBOC Combinatorial Small Molecule Libraries,” J. Comb Chern. 2009, 11(1), 91-102.
BD Life Sciences, 2018, BD AbSeq antibody-oligo conjugates, www.bd.com/genomics, 2 pp.
BD Life Sciences, 2018, BD AbSeq on the BD Rhapsody system: Exploration of single-cell gene regulation by simultaneous digital mRNA and protein quantification, www.bd.com/genomics, 7 pp.
Bontoux et al., “Integrating whole transcriptome assays on a lab-on-a-chip for single cell gene profiling”, Lab on a Chip 2008, 8(3), 443-450.
Bose et al., “Scalable microfluidics for single-cell RNA printing and sequencing,” Genome Biology 2015, 16(120), 1-16.
Brady et al., “Construction of cDNA libraries form single cells”, Methods in Enzymology 1993, (225), 611-623.
Brinza et al., “Detection of somatic mutations at 0.1% frequency from cfDNA in peripheral blood with a multiplex next-generation sequencing assay,” Conference Poster, AACR 107th Annual Meeting, Apr. 16-20, 2016, 1 p.
Buggenum et al., “A covalent and cleavable antibody DNA conjugation strategy for sensitive protein detection via immunoPCR,” Scientific Reports 2016, 6(22675), 1-12.
Buschmann et al., Enhancing the detection of barcoded reads in high throughput DNA sequencing DNA by controlling the false discovery rate, BMC Bioinformatics, 15(1), 264, 1-16.
Bustin, “Absolute quantification of mRNA using real-time reverse transcription polymerase chain reaction assays,” Journal of Molecular Endocrinology 2000, 25, 169-193.
Cao et al., “Comprehensive single-cell transcriptional profiling of a multicellular organism,” Science 2017, 357, 661-667.
Caruccio et al., “Nextera (TM) Technology for NGS DNA Library Preparation: Simultaneous Fragmentation and Tagging by in Vitro Transposition,” EpiBio 2009, 16(3), 4-6.
Chapin et al., “Rapid microRNA Profiling on Encoded Gel Microparticles,” Angew Chem Int Ed Engl. 2011, 50(10), 2289-2293.
Civil Cover Sheet filed Nov. 15, 2018 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 1 pp.
Clontech Laboratories, Inc., “Smart™ PCR cDNA Synthesis Kit User Manual,” Clontech 2007, 1-39.
Cloonan et al., “Stem cell transcriptome profiling via massive-scale mRNA sequencing”, Nature Methods 2008, 5(7), 613-619.
Communication of a Notice of Opposition dated Jul. 27, 2016 in European Patent Application No. EP 10762102.1.
Complaint filed in Becton, Dickinson and Company and Cellular Research Inc. v. 10X Genomics, Inc. dated Nov. 15, 2018 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 141 pp.
Costa et al., “Single-Tube Nested Real-Time PCR as a New Highly Sensitive Approach to Trace Hazelnut,” Journal of Agricultural and Food Chemistry 2012, 60, 8103-8110.
Cotten et al., “Selection of proteins with desired properties from natural proteome libraries using mRNA display,” Nature Protocols 2011, 6, 1163-1182.
Custom Antibody Services, Precision Antibody, accessed Apr. 16, 2014, 2 pp.
Day et al., “Immobilization of polynucleotides on magnetic particles,” Biochem. J. 1991, 278, 735-740.
Decision of Refusal dated Aug. 21, 2017 in Japanese Patent Application No. 2014-558975.
Defendant 10X Genomic Inc.'s Notice of Service for Initial Requests for Production and Interrogatories Served to Becton, Dickinson, and Company and Cellular Research, Inc., dated May 31, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 2 pp.
Defendant 10X Genomics Inc's, Notice of Service of Technical Documents, dated Jul. 8, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 2 pp.
Defendant 10X Genomic's Motion for Admission Pro Hac Vice of Paul Ehrlich, Azra Hadzimehmedovic and Aaron Nathan, Pursuant to Local Rule 83.5, dated May 1, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 5 pp.
Defendant 10X Genomic's Notice of Service for Initial Disclosures served to Opposing Counsel, dated Jun. 7, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 2 pp.
Defendant 10X Genomic's Request for Oral Argument Under D. Del. LR 7.1.4, dated Apr. 18, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA 2 pp.
Defendant 10X Genomic's Response Letter to Judge Richard G. Andrews re Request for a Rule 16, dated Apr. 16, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 2 pp.
Defendant 10X Genomics, Inc.'s [Proposed] Order for Partial Dismissal Pursuant to Federal Rules of Civil Procedure 12(b)(6), dated Jan. 18, 2019 in the USDC District of Delaware, C.A. No. 18-1800-RGA, 1 pp.
Defendant 10X Genomics, Inc.'s Letter to Judge Andrews in Response to Plaintiff's Letter of Supplemental Authority, dated Jul. 11, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 2 pp.
Defendant 10X Genomics, Inc.'s Motion for Admission Pro Hac Vice Pursuant to Local Rule 83.5, dated Jan. 18, 2019 in the USDC District of Delaware, C.A. No. 18-1800-RGA, 5 pp.
Defendant 10X Genomics, Inc.'s Motion to Dismiss Pursuant to Federal Rule of Civil Procedure 12(b)(6), dated Jan. 18, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 1 pp.
Defendant 10X Genomics, Inc.'s Motion to Dismiss the First Amended Complaint Pursuant to Federal Rule of Civil Procedure 12(b)(6), dated Mar. 1, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 1 pp.
Defendant 10X Genomics, Inc.'s Opening Brief in Support of Its Motion to Dismiss Pursuant to Federal Rule of Civil Procedure 12(b)(6), dated Jan. 18, 2019 in the USDC District of Delaware, C.A. No. 1:18-cv-01800-RGA, 25 pp.
Defendant 10X Genomics, Inc.'s Opening Brief in Support of Its Motion to Dismiss Pursuant to Federal Rule of Civil Procedure 12(b)(6), dated Mar. 1, 2019 in the USDC District of Delaware, C.A. No. 18-1800 RGA, 26 pp.
Defendant 10X Genomics, Inc.'s Rule 7.1 Disclosure Statement, dated Jan. 18, 2019 in the USDC District of Delaware, C.A. No. 18-1800-RGA, 1 pp. 1.
Defendant 10X Genomics, Inc's Proposed Order for Dismissal pursuant to Federal Rules of Civil Procedure 12(b)(6), filed Mar. 1, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 1 pp.
Defendant 10X Genomics's Reply Brief in support of its Motion to Dismiss Pursuant to Federal Rule of Civil Procedure 12(b)(6), dated Apr. 12, 2019 in USDC District of Delaware, C.A. No. 18-1800 RGA, 15 pp.
Delley et al., “Combined aptamer and transcriptome sequencing of single cells,” bioRxiv 2017, 1-10.
Eberwine et al., “Analysis of gene expression in single live neurons,” Proc. Natl. Acad. Sci. 1992, 89, 3010-3014.
Evanko et al., “Hybridization chain reaction,” Nature Methods 2004, 1(3), 186-187.
Examination Report dated Apr. 26, 2019 in European Patent Application No. 16710357.1.
Examination Report dated Aug. 2, 2019 in European Patent Application No. 17202409.3.
Examination Report dated Dec. 12, 2018 in European Patent Application No. 16719706.0.
Examination Report dated Dec. 4, 2019 in European Patent Application No. 16719706.0.
Examination Report dated Feb. 6, 2019 in European Patent Application No. 13754428.4.
Examination Report dated Jan. 2, 2019 in European Patent Application No. 16757986.1.
Examination Report dated Jan. 3, 2018 in UK Patent Application No. 1609740.4.
Examination Report dated Jul. 20, 2018 in Australian Patent Application No. 2014312208.
Examination Report dated May 12, 2020 in Australian Patent Application No. 2018220004.
Examination Report dated Jul. 24, 2019 in European Patent Application No. 16714081.3.
Examination Report dated Jun. 18, 2019 in European Patent Application No. 16710551.9.
Examination Report dated Mar. 16, 2018 in European Patent Application No. 13754428.4.
Examination Report dated Mar. 18, 2019 in Singapore Patent Application No. 11201405274W.
Examination Report dated Oct. 10, 2017 in European Patent Application No. 14761937.3.
Examination Report dated Oct. 11, 2019 in European Patent Application No. 16757986.1.
Examination Report dated Oct. 24, 2017 in Australian Patent Application No. 2013226081.
Examination Report dated Sep. 26, 2018 in European Patent Application No. 16714081.3.
Examination Report dated Sep. 5, 2018 in European Patent Application No. 16710357.1.
Examination Report dated Feb. 19, 2020 in European Patent Application No. 16710551.9.
Examination Report dated Mar. 18, 2020 in European Patent Application No. 17202409.3.
Exhibit A filed Jul. 10, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 25 pp.
Exhibits 12-32 filed Feb. 8, 2019 in the USDCfor the District of Delaware, C.A. 18-1800-RGA, 795 pp.
Exhibits A-D filed Jan. 18, 2019 in the USDC District of Delaware, C.A. No. 1:18-cv-01800-RGA, 47 pp.
Exhibits A-E filed Mar. 1, 2019 in the USDC District of Delaware, C.A. No. 18-1800 RGA, 75 pp.
Extended European Search Report dated Feb. 8, 2018 in European Patent Application No. 17202409.3.
Extended European Search Report dated Jun. 11, 2018 in European Patent Application No. 16740872.3.
Extended European Search Report dated Mar. 22, 2019 in European Patent Application No. 18195513.9.
Final Office Action dated Apr. 22, 2019 in U.S. Appl. No. 15/987,851.
Final Office Action dated Dec. 4, 2019 in U.S. Appl. No. 15/596,364.
Final Office Action dated Feb. 19, 2019 in U.S. Appl. No. 14/381,526.
Final Office Action dated Feb. 4, 2020 in U.S. Appl. No. 15/715,028.
Final Office Action dated Jan. 16, 2020 in U.S. Appl. No. 16/012,584.
Final Office Action dated Jan. 25, 2018 in U.S. Appl. No. 14/381,526.
Final Office Action dated Jan. 29, 2020 in U.S. Appl. No. 14/381,488.
Final Office Action dated Jan. 8, 2020 in U.S. Appl. No. 15/459,977.
Final Office Action dated Jul. 20, 2018 in U.S. Appl. No. 15/217,886.
Final Office Action dated Jul. 5, 2018 in U.S. Appl. No. 15/004,618.
Final Office Action dated Mar. 1, 2019 in U.S. Appl. No. 16/012,584.
Final Office Action dated May 10, 2018 in U.S. Appl. No. 14/381,488.
Final Office Action dated May 2, 2019 in U.S. Appl. No. 16/012,635.
Final Office Action dated May 3, 2018 in U.S. Appl. No. 15/046,225.
Final Office Action dated May 3, 2019 in U.S. Appl. No. 15/937,713.
Final Office Action dated Nov. 16, 2017 in U.S. Appl. No. 14/381,488.
Final Office Action dated Nov. 16, 2018 in U.S. Appl. No. 15/134,967.
Final Office Action dated Oct. 16, 2017 in U.S. Appl. No. 15/409,355.
Final Office Action dated Oct. 2, 2019 in U.S. Appl. No. 15/084,307.
Final Office Action dated Sep. 18, 2019 in U.S. Appl. No. 15/055,407.
Final Office Action dated Mar. 9, 2020 in U.S. Appl. No. 15/987,851.
Final Office Action dated Apr. 28, 2020 in U.S. Appl. No. 15/134,967.
First Action Interview Office Action Summary dated Jan. 25, 2019 in U.S. Appl. No. 15/987,851.
First Action Interview Pilot Program Pre-Interview Communication dated Oct. 15, 2018 in U.S. Appl. No. 15/987,851.
Flanigon et al., “Multiplex protein detection with DNA readout via mass spectrometry,” N Biotechnol. 2013, 30(2), 153-158.
Forster et al., “A human gut bacterial genome and culture collection for improved metagenomic analyses,” Nature Biotechnology 2019, 37, 186-192.
Fu et al., “Molecular indexing enables quantitative targeted RNA sequencing and reveals poor efficiencies in standard library preparation,” PNAS 2014, 111(5), 1891-1896.
Gong et al., “Simple Method Prepare Oligonucleotide-Conjugated Antibodies and Its Application in Multiplex Protein Detection in Single Cells,” Bioconjugate Chem. 2016, 27, 217-225.
Gu et al., “Complete workflow for detection of low frequency somatic mutations from cell-free DNA using Ion Torrent™ platforms,” Conference Poster, AACR 107th Annual Meeting, Apr. 16-20, 2016, 1 p.
Han et al., “An approach to multiplexing an immunosorbent assay with antibody-oligonucleotide conjugates,” Bioconjug Chem. 2010, 21(12), 2190-2196.
Harbers, “The current status of cDNA cloning,” Genomics 2008, 91, 232-242.
Hartmann, “Gene expression profiling of single cells on large-scale oligonucleotide arrays”, Nucleic Acids Research, (Oct. 2006) vol. 34, No. 21, p. e143, 1-12.
Holcomb et al., “Abstract 1853: Single-cell multiplexed profiling of protein-level changes induced by EGFR inhibitor gefitinib,” Cancer Res 2016, 76(14 Suppl), Abstract 1853.
How many species of bacteria are there? Wisegeek.org, accessed Jan. 21, 2014, 2 pp.
Hu et al., “Dissecting Cell-Type Composition and Activity-Dependent Transcriptional State in Mammalian Brains by Massively Parallel Single-Nucleus RNA-Seq,” Molecular Cell 2017, 68, 1006-1015.
Hu et al., “Single Cell Multi-Omics Technology: Methodology and Application,” Frontiers in Cell and Developmental Biology 2018, 6(28), 1-13.
International Preliminary Report on Patentability dated Aug. 15, 2019 in PCT Application No. PCT/US2018/014385.
International Preliminary Report on Patentability dated Mar. 26, 2019 in PCT Application No. PCT/US2017/053331.
International Search Report and Written Opinion dated Dec. 4, 2019 in PCT Application No. PCT/US2019/053868.
International Search Report and Written Opinion dated Jan. 27, 2020 in PCT Application No. PCT/US2019/048179.
International Search Report and Written Opinion dated Jul. 16, 2018 in PCT Application No. PCT/US2018/024602.
International Search Report and Written Opinion dated Jun. 24, 2019 in PCT Application No. PCT/US2019/030175.
International Search Report and Written Opinion dated Mar. 20, 2018 in PCT Application No. PCT/US2017/053331.
International Search Report and Written Opinion dated Nov. 27, 2019 in PCT Application No. PCT/US2019/046549.
International Search Report and Written Opinion dated Oct. 16, 2019 in PCT Application No. PCT/US2019/030245.
International Search Report and Written Opinion dated Oct. 8, 2019 in PCT Application No. PCT/US2019/043949.
International Search Report and Written Opinion dated Sep. 8, 2017 in PCT Application No. PCT/US2017/030097.
International Search Report and Written Opinion dated Mar. 30, 2020 in PCT Application No. PCT/US2019/060243.
International Search Report and Written Opinion dated Mar. 30, 2020 in PCT Application No. PCT/US2019/065237.
Invitation to Pay Fees dated May 16, 2018 in PCT Application No. PCT/US2018/024602.
Invitation to Pay Fees dated Nov. 26, 2019 in PCT Application No. PCT/US2019/048179.
Invitation to Pay Fees dated May 7, 2020 in PCT Application No. PCT/US2020/017890.
Islam et al., “Highly multiplexed and strand specific single-cell RNA 5′ end sequencing,” Nature Protocols 2012, 7(5), 813-828.
Jason J. Rawnsley of Richards, Layton and Finger, P.A., Entry of Appearance dated Jan. 18, 2019 in the USDC District of Delaware, C.A. No. 18-1800-RGA, 1 pp.
Jiang et al., “Synthetic spike-in standards for RNA-seq experiments,” Genome Res. 2011, 21, 1543-1551.
Joint Stipulation and Order to Extend Time to Respond to Plaintiff's First Amended Complaint, dated Feb. 21, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 2 pp.
Joint Stipulation and Order to Extended Time to Submit Agreed Document Production Protocol, filed Jun. 28, 2019 in the USDC for the District of Delaware, C.A. 18-1800 (RGA), 1 pp.
Joint Stipulation and Order to Request Extended Time to File Opposition to Defendant's Motion to Dismiss dated, Mar. 8, 2019 in the USDC District of Delaware, C.A. No. 18-1800 RGA, 2 pp.
Joint Stipulation and Order to Request Extended Time to Submit a proposed Protective Order, dated Jun. 7, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 1 pp.
Joint Stipulation and Order to Request Extended Time to Submit Agreed Document Production Protocol, dated Jul. 11, 2019 in the USDC for the District of Delaware, C.A. 18-1800 (RGA), 1 pp.
Kang et al., “Targeted sequencing with enrichment PCR: a novel diagnostic method for the detection of EGFR mutations,” Oncotarget 2015, 6(15), 13742-13749.
Karrer et al., “In situ isolation of mRNA from individual plant cells: creation of cell-specific cDNA libraries,” Proc. Natl. Acad. Sci. USA 1995, 92, 3814-3818.
Kausch et al., “Organelle Isolation by Magnetic Immunoabsorption,” BioTechniques 1999, 26(2), 336-343.
Kirsebom et al., “Stimuli-Responsive Polymers in the 21st Century: Elaborated Architecture to Achieve High Sensitivity, Fast Response, and Robust Behavior,” Journal of Polymer Science: Part B: Polymer Physics 2011, 49, 173-178.
Ko et al., “RNA-conjugated template-switching RT-PCR method for generating an Escherichia coli cDNA library for small RNAs,” Journal of Microbiological Methods 2006, 64, 297-304.
Kozlov et al., “A high-complexity, multiplexed solution-phase assay for profiling protease activity on microarrays,” Comb Chem High Throughput Screen 2008, 11(1), 24-35.
Kurimoto et al., “Global single-cell cDNA amplification to provide a template for representative high-density oligonucleotide microarray analysis,” Nature Protocols 2007, 2(3), 739-752.
Lass-Napiorkowska et al., “Detection methodology based on target molecule-induced sequence-specific binding to a single-stranded oligonucleotide,” Anal Chem. 2012, 84(7), 3382-3389.
Lee et al., “Universal process-inert encoding architecture for polymer microparticles,” Nature Materials 2014, 13(5), 524-529.
Letter to Judge Andrews regarding Agreement on Proposed Scheduling Order, dated May 7, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 1 pp.
Letter to Judge Andrews regarding Notice of Supplemental Authority, dated Jul. 10, 2019 in the USDC for the District of Delaware, C.A. 18-1800(RGA), 2pp.
Letter to Judge Richard G. Andrews Requesting a Rule 16 Conference, dated Apr. 15, 2019 in the USDC for the District of Delaware, C.A. 18-1800 (RGA), 1 pp.
Lin et al., “Self-Assembled Combinatorial Encoding Nanoarrays for Multiplexed Biosensin,” Nano Lett. 2007, 7(2), 507-512.
Lundberg et al., “Practical innovations for high-throughput amplicon sequencing,” Nature Methods 2013, 10(10), 999-1007.
Lundberg et al., “Supplementary Information for: Practical innovations for high-throughput amplicon sequencing,” Nature Methods 2013, 1-24.
Macaulay et al., “Single Cell Genomics: Advances and Future Perspectives,” PLoS Genetics 2014, 10(1), 1-9.
Maeda et al., “Development of a DNA barcode tagging method for monitoring dynamic changes in gene expression by using an ultra high-throughput sequencer,” BioTechniques 2008, 45(1), 95-97.
Mardis, “Next-generation DNA sequencing methods”, Annu. Rev. Genomics Hum. Genet. 2008, 9, 387-402.
Marguerat et al., “Next-generation sequencing: applications beyond genomes,” Biochem. Soc. Trans. 2008, 36(5), 1091-1096.
Massachusetts General Hospital, Overview of Illumina Chemistry, http://nextgen.mgh.harvard.edu/IlluminaChemistry.html, downloaded Jan. 28, 2020, 2 pp.
Meyer et al., “Parallel tagged sequencing on the 454 platform,” Nature Protocols 2008, 3(2), 267-278.
Motion and Order for Admission Pro Hac Vice Pursuant to Local Rule 83.5, dated Jan. 24, 2019 in the USDC District of Delaware, C.A. No. 18-1800-RGA, 7 pp.
Non-Final Office Action dated Aug. 20, 2019 in U.S. Appl. No. 15/715,028.
Non-Final Office Action dated Jan. 12, 2018 in U.S. Appl. No. 15/217,886.
Non-Final Office Action dated Jan. 14, 2019 in U.S. Appl. No. 16/219,553.
Non-Final Office Action dated Jan. 17, 2020 in U.S. Appl. No. 15/084,307.
Non-Final Office Action dated Jan. 7, 2019 in U.S. Appl. No. 15/055,407.
Non-Final Office Action dated Jan. 9, 2018 in U.S. Appl. No. 15/217,896.
Non-Final Office Action dated Jul. 25, 2018 in U.S. Appl. No. 15/108,268.
Non-Final Office Action dated Jul. 9, 2019 in U.S. Appl. No. 15/596,364.
Non-Final Office Action dated Jun. 17, 2019 in U.S. Appl. No. 14/381,488.
Non-Final Office Action dated Jun. 2, 2017 in U.S. Appl. No. 14/381,526.
Non-Final Office Action dated Mar. 19, 2019 in U.S. Appl. No. 15/046,225.
Non-Final Office Action dated Mar. 8, 2018 in U.S. Appl. No. 15/608,780.
Non-Final Office Action dated May 15, 2019 in U.S. Appl. No. 15/084,307.
Non-Final Office Action dated May 23, 2019 in U.S. Appl. No. 15/459,977.
Non-Final Office Action dated Nov. 1, 2017 in U.S. Appl. No. 15/667,125.
Non-Final Office Action dated Nov. 26, 2018 in U.S. Appl. No. 15/937,713.
Non-Final Office Action dated Nov. 29, 2019 in U.S. Appl. No. 15/937,713.
Non-Final Office Action dated Nov. 5, 2018 in U.S. Appl. No. 16/038,790.
Non-Final Office Action dated Nov. 9, 2017 in U.S. Appl. No. 15/004,618.
Non-Final Office Action dated Oct. 25, 2018 in U.S. Appl. No. 16/012,584.
Non-Final Office Action dated Oct. 4, 2018 in U.S. Appl. No. 15/260,106.
Non-Final Office Action dated Sep. 18, 2019 in U.S. Appl. No. 16/194,819.
Non-Final Office Action dated Sep. 8, 2017 in U.S. Appl. No. 15/046,225.
Non-Final Office Action dated Sep. 8, 2017 in U.S. Appl. No. 15/134,967.
Non-Final Office Action dated Mar. 17, 2020 in U.S. Appl. No. 15/055,407.
Non-Final Office Action dated Mar. 12, 2020 in U.S. Appl. No. 16/789,358.
Non-Final Office Action dated Mar. 26, 2020 in U.S. Appl. No. 16/789,311.
Non-Final Office Action dated Mar. 26, 2020 in U.S. Appl. No. 16/012,635.
Notice of Allowance dated Dec. 27, 2019 in U.S. Appl. No. 15/260,106.
Notice of Allowance dated Jan. 9, 2019 in U.S. Appl. No. 15/603,239.
Notice of Allowance dated Mar. 21, 2019 in U.S. Appl. No. 15/993,468.
Notice of Allowance dated May 28, 2019 in U.S. Appl. No. 16/219,553.
Notice of Allowance dated Mar. 20, 2019 in U.S. Appl. No. 16/219,553.
Notice of Allowance dated Nov. 11, 2019 in Japanese Patent Application No. 2017-245295.
Notice of Allowance dated Nov. 29, 2019 in U.S. Appl. No. 16/012,635.
Notice of Allowance dated Sep. 24, 2019 in U.S. Appl. No. 15/217,886.
Notice of Allowance dated Mar. 5, 2020 in U.S. Appl. No. 15/217,886.
Notice of Allowance dated Mar. 27, 2020 in U.S. Appl. No. 15/596,364.
Notice of Allowance dated Mar. 30, 2020 in U.S. Appl. No. 15/937,713.
Notice of Allowance dated Apr. 15, 2020 in U.S. Appl. No. 16/012,635.
Notice of Reason for Refusal dated Nov. 21, 2019 in Korean Patent Application No. 10-2016-7008144.
Notice of Reasons for Rejection dated Apr. 2, 2018 in Japanese Patent Application No. 2014-558975.
Notice of Reasons for Rejection dated Aug. 31, 2018 in Japanese Patent Application No. 2016-520632.
Notice of Reasons for Rejection dated Dec. 5, 2018 in Japanese Patent Application No. 2017-245295.
Notice of Reasons for Rejection dated Feb. 25, 2020 in Japanese Patent Application No. 2019-014564.
Notice of Reasons for Refusal dated May 11, 2020 in Japanese Patent Application No. 2017-549390.
Notice of Service of Disclosures to Opposing Counsel, dated Jun. 10, 2019 in the USDC for the District of Delaware, C.A. 18-1800 (RGA), 3 pp.
Notice of Service of Interrogatories and First Request of Documents and Things to Defendant 10X Genomics, Inc., dated Jul. 5, 2019 in the USDC for the District of Delaware, C.A. 18-1800 (RGA), 3 pp.
Notice, Consent, and Reference of a Civil Action to a Magistrate Judge (Rule 73.1), filed Nov. 15, 2018 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 3 pp.
Notification Prior to Examination dated Nov. 27, 2019 in Israeli Patent Application No. 265478.
Novak et al., “Single-Cell Multiplex Gene Detection and Sequencing with Microfluidically Generated Agarose Emulsions,” Angew. Chem. Int. Ed. 2011, 50, 390-395.
Office Action dated Dec. 13, 2018 in Canadian Patent Application No. 2,865,575.
Office Action dated Dec. 19, 2017 in Chinese Patent Application No. 201480061859.1.
Office Action dated Feb. 15, 2018 in Canadian Patent Application No. 2,865,575.
Office Action dated Jan. 2, 2019 in Chinese Patent Application No. 201480059505.3.
Office Action dated Sep. 7, 2018 in Chinese Patent Application No. 201480061859.1.
Office Action dated Mar. 4, 2020 in Canadian Patent Application No. 2,865,575.
Opposition to Defendant's Motion to Dismiss Pursuant to Federal Rule of Civil Procedure 12(b)(6) dated Feb. 15, 2019, in the USDC for the District of Delaware, C.A. 18-800-RGA, 3 pp.
Oral Order by Judge Andrews Canceling Scheduling Conference set for May 8, 2019.
Order Scheduling ADR Mediation Teleconference, filed May 13, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 4pp.
Order Setting Rule 16(b) Conference as Ordered by Judge Andrews Pursuant to Fed. R. Civ. P. 16(b), ruling dated Apr. 17, 2019 in the USDC District of Delaware, C.A. 18-1800-RGA, 1 pp.
Ozkumur et al., “Inertial Focusing for Tumor Antigen-Dependent and -Independent Sorting of Rare Circulating Tumor Cells,” Science Translational Medicine 2013, 5(179), 1-20.
Pérez-Rentero et al., “Synthesis of Oligonucleotides Carrying Thiol Groups Using a Simple Reagent Derived from Threoninol,” Molecules 2012, 17, 10026-10045.
Peterson et al., “Multiplexed quantification of proteins and transcripts in single cells,” Nature Biotechnology 2017, 35, 936-939.
Pfaffl et al., “Determination of stable housekeeping genes, differentially regulated target genes and sample integrity: BestKeeper—Excel-based tool using pair-wise correlations,” Biotechnology Letters, 26(6), 505-515.
Picelli et al., “Single-cell RNA-sequencing: The future of genome biology is now,” RNA Biology 2017, 14(5), 637-650.
Plaintiff's Brief in Opposition to Defendant's Motion to Dismiss Pursuant to Fed. R. Civ. P. 12(b)(6), filed Mar. 29, 2019 in the USDC District of Delaware, C.A. No. 18-1800 (RGA), 27 pp.
Plaintiff's First Amended Complaint filed on Feb. 8, 2019, in the USDC for the District of Delaware, C.A. 18-1800-RGA, 178 pp.
Pre-interview communication dated Nov. 27, 2018 in U.S. Appl. No. 16/012,635.
Preissl et al., “Single-nucleus analysis of accessible chromatin in developing mouse forebrain reveals cell-type-specific transcriptional regulation,” Nature Neuroscience 2018, 21(3), 432-439.
Proposed Stipulated Protective Order Pursuant to Rule 26(c) of the Federal Rules of Civil Procedure, filed Jun. 20, 2019 In the USDC for the District of Delaware, C.A. 18-1800 (RGA), 26 pp.
Report on the Filing or Determination of an Action Regarding a Patent or Trademark filed Nov. 15, 2018 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 2 pp.
Restriction Requirement dated Jun. 19, 2019 in U.S. Appl. No. 15/596,364.
Restriction Requirement dated Mar. 29, 2019 in U.S. Appl. No. 15/715,028.
Rule 7.1 Disclosure Statement dated Nov. 15, 2018 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 1 pp.
Sah et al., “Complete Genome Sequence of a 2019 Novel Coronavirus (SARS-CoV-2) Strain Isolated in Nepal,” Microbiol Resour Announc. 2020, 9(11), e00169-20, 3 pp.
Sano et al., “Immuno-PCR: Very Sensitive Antigen Detection by Means of Specific Antibody-DNA Conjugates,” Science 1992, 258, 120-122.
Scheduling Order pursuant to Local Rule 16.1(b), filed May 7, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 10 pp.
Scheduling Order Signed by Judge Andrews, dated May 8, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 10 pp.
Search Report and Written Opinion dated Jan. 26, 2016 in Singapore patent application No. 1120140527W.
Shahi et al., “Abseq: ultrahigh-throughput single cell protein profiling with droplet microfluidic barcoding,” Scientific Reports 2017, 7(44447), 1-10.
Shendure et al., “Next-generation DNA sequencing,” Nature Biotechnology 2008, 26(10), 1135-1145.
Shortreed et al., “A thermodynamic approach to designing structure-free combinatorial DNA word sets,” Nucleic Acids Res. 2005, 33(15), 4965-4977.
Shum et al., “Quantitation of mRNA Transcripts and Proteins Using the BD Rhapsody™ Single-Cell Analysis System,” Adv Exp Med Biol. 2019,1129, 63-79.
Soares et al., “Construction and characterization of a normalized cDNA library,” Proc. Natl., Acad. Sci. 1994, 91, 9228-9232.
Sogin et al., “Microbial diversity in the deep sea and the underexplored “rare biosphere”,” PNAS 2008, 103(32), 12115-12120.
Sommer et al., “Minimal homology requirements for PCR primers,” Nucleic Acids Research 1989, 17(16), 6749.
Song et al., “Design rules for size-based cell sorting and sheathless cell focusing by hydrophoresis,” Journal of Chromatography A 2013, 1302, 191-196.
Statement of Opposition dated Jul. 21, 2016 filed against European Patent No. EP2414548B1.
Statement of Opposition filed against European Patent No. EP2414548B1 on Jul. 26, 2016.
Statement of Opposition of Strawman Limited filed against European Patent No. EP2414548B1 on Jul. 19, 2016.
Statement regarding Third-Party Submission filed on Jun. 6, 2018 for U.S. Appl. No. 15/847,752.
Stipulated Protective Order Pursuant to Rule 26(c) of the Federal Rules of Civil Procedure, dated Jun. 21, 2019 in the USDC for the District of Delaware, C.A. 18-1800 (RGA), 26 pp.
Stipulation and Order to Extend Time to File Opposition to Motion to Dismiss, and Reply in Support of the Motion, dated Jan. 28, 2019 in the USDC for the District of Delaware, C.A. 18-1800-RGA, 2 pp.
Stoeckius et al., “Large-scale simultaneous measurement of epitopes and transcriptomes in single cells,” Nature Methods 2017, 14(9), 865-868.
Subkhankulova et al., “Comparative evaluation of linear and exponential amplification techniques for expression profiling at the single cell level,” Genome Biology 2006, 7(3), 1-16.
Submission dated Jan. 15, 2018 in preparation for upcoming oral proceedings in opposition against European Patent No. EP2414548B1.
Summons in a Civil Action to Defendant 10X Genomics, Inc. filed Nov. 16, 2018 in the USDC for the District of Delaware, Civil Action No. 18-1800, 2 pp.
Sun et al., “Ultra-deep profiling of alternatively spliced Drosophila Dscam isoforms by circularization-assisted multi-segment sequencing,” EMBO J. 2013, 32(14), 2029-2038.
Tang et al., “RNA-Seq analysis to capture the transcriptome landscape of a single cell,” Nature Protocols 2010, 5(3), 516-535.
Third-Party Submission filed on May 21, 2018 for U.S. Appl. No. 15/847,752.
Ullal et al., “Cancer cell profiling by barcoding allows multiplexed protein analysis in fine needle aspirates,” Sci Transl Med. 2014, 6(219), 22 pp.
Unopposed Motion to Extend Time for Defendant's Response, dated Dec. 4, 2018 in the USDC for the District of Delaware, C.A. 18-1800-(RGA), 2 pp.
Vollbrecht et al., “Validation and comparison of two NGS assays for the detection of EGFR T790M resistance mutation in liquid biopsies of NSCLC patients,” Oncotarget 2018, 9(26), 18529-18539.
Wang et al., “Combining Gold Nanoparticles with Real-Time Immuno-PCR for Analysis of HIV p24 Antigens,” Proceedings of ICBBE 2007, 1198-1201.
Weibrecht et al., “Proximity ligation assays: a recent addition to the proteomics toolbox,” Expert Rev. Proteomics 2010, 7(3), 401-409.
Zagordi et al., “Error correction of next-generation sequencing data and reliable estimation of HIV quasispecies,” Nucleic Acids Research 2010, 38(21), 7400-7409.
Zhou et al., “Photocleavable Peptide-Oligonucleotide Conjugates for Protein Kinase Assays by MALDI-TOF MS,” Mol. BioSyst. 2012, 8, 2395-2404.
Zhu et al., “Reverse Transcriptase Template Switching: A Smart Approach for Full-Length cDNA Library Construction,” BioTechniques 2001, 30(4), 892-897.
Advisory Action dated Aug. 25, 2020 in U.S. Appl. No. 15/084,307.
Biosciences Product Catalogue, Dynal® Catalog 1999, Oslo, Norway, 49-51.
Defendant 10X Genomics Reply Brief in Support of its Motion to Dismiss Pursuant to Federal Rule of Civil Procedure 12(b)(6), dated Apr. 12, 2019 in the USDC for the District of Delaware, C.A. No. 18-1800-RGA, 15 pp.
Dube et al., “Mathematical Analysis of Copy Number Variation in a DNA Sample Using Digital PCR on a Nanofluidic Device,” PLoS One 2008, 3(8) e2876.
Examination Report dated Jul. 6, 2020 in European Patent Application No. 17781265.8.
Examination Report dated Sep. 21, 2020 in European Patent Application No. 18703156.2.
Final Office Action dated Jun. 5, 2020 in U.S. Appl. No. 15/084,307.
Final Office Action dated Sep. 14, 2020 in U.S. Appl. No. 16/789,358.
Final Office Action dated Sep. 22, 2020 in U.S. Appl. No. 16/789,311.
Final Office Action dated Sep. 25, 2020 in U.S. Appl. No. 15/055,407.
Final Office Action dated Dec. 7, 2020 in U.S. Appl. No. 16/012,584.
International Preliminary Report on Patentability dated Nov. 3, 2020 in PCT Application No. PCT/US2019/030175.
International Preliminary Report on Patentability dated Nov. 3, 2020 in PCT Application No. PCT/US2019/030245.
International Search Report and Written Opinion dated May 18, 2020 in PCT Application No. PCT/US2020/014339.
International Search Report and Written Opinion dated Jun. 30, 2020 in PCT Application No. PCT/US2020/017890.
International Search Report and Written Opinion dated Nov. 12, 2020 in PCT Application No. PCT/US2020/042880.
Kang et al., “Application of multi-omics in single cells,” Ann Biotechnol. 2018, 2(1007), 1 -8.
Kooiker & Xue, “cDNA Library Preparation,” Cereal Genomics 2013, 1099, 29-40.
Kozarewa & Turner, “96-Plex Molecular Barcoding for the Illumina Genome Analyzer,” High-Throughput Next Generation Sequencing. Methods in Molecular Biology (Methods and Applications) 2011, 733, 24 pp. DOI: 10.1007/978-1-61779-089-8_20.
Non-Final Office Action dated Aug. 20, 2019 for U.S. Appl. No. 15/715,028.
Non-Final Office Action dated Jun. 8, 2020 in U.S. Appl. No. 15/715,028.
Non-Final Office Action dated Aug. 4, 2020 in U.S. Appl. No. 15/459,977.
Non-Final Office Action dated Aug. 19, 2020 in U.S. Appl. No. 16/374,626.
Non-Final Office Action dated Aug. 25, 2020 in U.S. Appl. No. 14/381,488.
Non-Final Office Action dated Dec. 4, 2020 in U.S. Appl. No. 16/677,012.
Non-Final Office Action dated Dec. 9, 2020 in U.S. Appl. No. 16/788,743.
Notice of Allowance dated Sep. 23, 2020 in Korean Patent Application No. 10-2016-7008144.
Notice of Allowance dated Oct. 29, 2020 in U.S. Appl. No. 15/987,851.
Notice of Opposition dated Jul. 27, 2016 for European Patent Application No. 10762102.1.
Office Action dated Jun. 22, 2020 in Chinese Patent Application No. 201680007351.2.
Office Action dated Jun. 22, 2020 in Chinese Patent Application No. 201680007652.5.
Office Action dated Jun. 23, 2020 in Chinese Patent Application No. 2016800157452.
Office Action dated Jul. 20, 2020 in Japanese Patent Application No. 2018-512152.
Office Action dated Oct. 29, 2020 in Chinese Patent Application No. 2018800377201.
Office Action dated Nov. 12, 2020 in European Patent Application No. 18716877.8.
Office Action dated Dec. 3, 2020 in European Patent Application No. 16719706.0.
Raj et al., “Stochastic mRNA synthesis in mammalian cells,” PLoS Biol. 2006, 4(10) 1707-1719.
Raj et al., “Imaging individual mRNA molecules using multiple singly labeled probes,” Nature Methods 2008, 5(10), 877-879.
Raj et al., “Single-Molecule Approaches to Stochastic Gene Expression,” Annu Rev Biophys 2009, 38, 255-270.
Rhee et al., “Simultaneous detection of mRNA and protein stem cell markers in live cells,” BMC Biotechnology 2009, 9(30), 1-10.
Search Report and Written Opinion dated Aug. 26, 2020 in Singapore Patent Application No. 10201806890V.
S.H.Ko, “An ‘equalized cDNA library’ by the reassociation of short double-stranded cDNAs,” Nucleic Acids Res. 1990, 18(19), 5705-5711.
Summons to Attend Oral Proceedings dated Nov. 16, 2020 in European Patent Application No. 17202409.3.
Ahern, “Biochemical, Reagent Kits Offer Scientists Good Return on Investment,” The Scientist 1995, 9(15), in 5 pages.
Examination Report dated Mar. 25, 2021 in European Patent Application No. 17781265.8.
Extended European Search Report dated May 6, 2021 in European Patent Application No. 20207621.2.
Extended European Search Report dated May 28, 2021 in European Patent Application No. 20209777.0.
Final Office Action dated Feb. 11, 2021 in U.S. Appl. No. 15/134,967.
Final Office Action dated Mar. 16, 2021 in U.S. Appl. No. 15/715,028.
Final Office Action dated Mar. 25, 2021 in U.S. Appl. No. 16/374,626.
Final Office Action dated Jun. 15, 2021 in U.S. Appl. No. 15/084,307.
Final Office Action dated Jul. 15, 2021 in U.S. Appl. No. 16/836,750.
Final Office Action dated Aug. 10, 2021 in U.S. Appl. No. 16/012,584.
Final Office Action dated Aug. 27, 2021 in U.S. Appl. No. 15/055,407.
Fitzgerald and Grivel, “A Universal Nanoparticle Cell Secretion Capture Assay,” Cytometry Part A 2012, 83A(2), 205-211.
Gratton et al., “Cell-permeable peptides improve cellular uptake and therapeutic gene delivery of replication-deficient viruses in cells and in vivo,” Nature Medicine 2003, 9(3), 357-362.
International Search Report and Written Opinion dated Jan. 19, 2021 in PCT Application No. PCT/US2020/059419.
International Search Report and Written Opinion dated Apr. 9, 2021 in PCT Application No. PCT/US2021/013137.
International Search Report and Written Opinion dated Apr. 21, 2021 in PCT Application No. PCT/US2021/015571.
International Search Report and Written Opinion dated May 4, 2021 in PCT Application No. PCT/US2021/013109.
International Search Report and Written Opinion dated May 11, 2021 in PCT Application No. PCT/US2021/013748.
International Search Report and Written Opinion dated Jul. 15, 2021 in PCT Application No. PCT/US2021/019475.
International Search Report and Written Opinion dated Jul. 20, 2021 in PCT Application No. PCT/US2021/015898.
International Search Report and Written Opinion dated Aug. 30, 2021 in PCT Application No. PCT/US2021/035270.
Invitation to Pay Fees dated May 25, 2021 in PCT Application No. PCT/US2021/01598.
Invitation to Pay Additional Search Fees dated Sep. 8, 2021 in PCT Application No. PCT/US2021/032319.
Invitation to Provide Informal Clarification dated Jun. 9, 2021 in PCT Application No. PCT/US2021/019475.
Janeway et al., “Structural variation in immunoglobulin constant regions,” Immunology: The Immune System in Health and Disease 1999, 101-103.
Livingstone, “rRNA depletion, poly(A) enrichment, or exonuclease treatment?” Tebu-Bio Blog 2015, in 1 page.
New COVID-19 Variants, Centers for Disease Control and Prevention 2021, accessed Jan. 21, 2021, 3 pp.
Non-Final Office Action dated Jan. 19, 2021 in U.S. Appl. No. 16/836,750.
Non-Final Office Action dated Feb. 21, 2021 in U.S. Appl. No. 16/535,080.
Non-Final Office Action dated Feb. 25, 2021 in U.S. Appl. No. 15/055,407.
Non-Final Office Action dated Feb. 25, 2021 in U.S. Appl. No. 15/084,307.
Non-Final Office Action dated Mar. 29, 2021 in U.S. Appl. No. 16/789,358.
Non-Final Office Action dated Apr. 14, 2021 in U.S. Appl. No. 16/789,311.
Non-Final Office Action dated May 18, 2021 in U.S. Appl. No. 16/535,080.
Non-Final Office Action dated Jun. 9, 2021 in U.S. Appl. No. 16/588,405.
Non-Final Office Action dated Aug. 17, 2021 in U.S. Appl. No. 16/551,620.
Non-Final Office Action dated Aug. 19, 2021 in U.S. Appl. No. 16/781,814.
Non-Final Office Action dated Aug. 31, 2021 in U.S. Appl. No. 15/715,028.
Non-Final Office Action dated Sep. 1, 2021 in U.S. Appl. No. 16/789,358.
Non-Final Office Action dated Sep. 14, 2021 in U.S. Appl. No. 16/707,780.
Notice of Allowance dated Jan. 13, 2021 in U.S. Appl. No. 14/381,488.
Notice of Allowance dated Jan. 13, 2021 in U.S. Appl. No. 15/459,977.
Notice of Allowance dated Apr. 26, 2021 in Japanese Patent Application No. 2019-014564.
Notice of Allowance dated Jun. 10, 2021 in Chinese Patent Application No. 2018800377201.
Notice of Allowance dated Aug. 16, 2021 in Japanese Patent Application No. 2018-512152.
Notice of Allowance dated Sep. 10, 2021 in U.S. Appl. No. 16/535,080.
Novus Biologicals, “Fixation and Permeability in ICC IF,” Novus Biologicals 2021, 1-3.
Office Action dated Jan. 4, 2021 in Japanese Patent Application No. 2017-549390.
Office Action dated Jan. 6, 2021 in Chinese Patent Application No. 201680052330.2.
Office Action dated Jan. 14, 2021 in Japanese Patent Application No. 2019-014564.
Office Action dated Jan. 15, 2021 in Korean Patent Application No. 10-2020-7033213.
Office Action dated Jan. 26, 2021 in Chinese Patent Application No. 201680007351.2.
Office Action dated Feb. 4, 2021 in Canadian Patent Application No. 2,865,575.
Office Action dated Feb. 20, 2021 in Chinese Patent Application No. 201680022865.5.
Office Action dated Mar. 1, 2021 in Chinese Patent Application No. 201680007652.5.
Office Action dated Mar. 2, 2021 in Chinese Patent Application No. 2016800157452.
Office Action dated Mar. 8, 2021 in Japanese Patent Application No. 2018-512152.
Office Action dated Mar. 16, 2021 in Chinese Patent Application No. 2018800377201.
Office Action dated May 10, 2021 in Japanese Patent Application No. 2019-566787.
Office Action dated May 21, 2021 in Chinese Patent Application No. 201680007351.2.
Office Action dated Jul. 26, 2021 in Korean Patent Application No. 10-2019-7011635.
Office Action dated Jul. 28, 2021 in Korean Patent Application No. 10-2020-7033213.
Office Action dated Aug. 13, 2021 in Chinese Patent Application No. 2017800587991.
Office Action dated Aug. 27, 2021 in Chinese Patent Application No. 2016800076525.
O'Shea et al., “Analysis of T Cell Receptor Beta Chain CDR3 Size Using RNA Extracted from Formalin Fixed Paraffin Wax Embedded Tissue,” Journal of Clinical Pathology 1997, 50(10), 811-814.
Prevette et al., “Polycation-Induced Cell Membrane Permeability Does Not Enhance Cellular Uptake or Expression Efficiency of Delivered DNA,” Molecular Pharmaceutics 2010, 7(3), 870-883.
Pringle et al., “In Situ Hybridization Demonstration of Poly-Adenylated RNA Sequences in Formalin-Fixed Paraffin Sections Using a Biotinylated Oligonucleotide Poly d(T) Probe,” Journal of Pathology 1989, 158, 279-286.
Restriction Requirement dated May 5, 2021 in U.S. Appl. No. 16/400,886.
Restriction Requirement dated May 28, 2021 in U.S. Appl. No. 16/781,814.
Restriction Requirement dated Jun. 4, 2021 in U.S. Appl. No. 16/551,620.
Restriction Requirement dated Sep. 20, 2021 in U.S. Appl. No. 16/525,054.
Stoeckius et al., “Cell Hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics,” Genome Biology 2018, 19(224), 1-12.
Takara Bio, “SMARTer Human BCR IgG IgM H/K/L Profiling Kit User Manual,” Takara Bio USA Inc. 2019, 1-22.
TotalSeq™-A0251 anti-human Hashtag 1 Antibody, BioLegend®, Jul. 2018, 1-10.
Zeberg et al., “The major genetic risk factor for severe COVID-19 is inherited from Neanderthals,” Nature 2020, 587(7835), 1-13.
Adey et al., “Rapid, low-input, low-bias construction of shotgun fragment libraries by high-density in vitro transposition,” Genome Biology 2010, 11 (R19), in 17 pages.
Brouilette et al., “A Simple and Novel Method for RNA-seq Library Preparation of Single Cell cDNA Analysis by Hyperactive Tn5 Transposase,” Developmental Dynamics 2012, 241, 1584-1590.
Examination Report dated Oct. 8, 2021 in European Patent Application No. 18716877.8.
Examination Report dated Nov. 18, 2021 in European Patent Application No. 19724003.9.
Examination Report dated Nov. 24, 2021 in European Patent Application No. 19762517.1.
Examination Report dated Dec. 6, 2021 in European Patent Application No. 18703156.2.
Examination Report dated Dec. 9, 2021 in European Patent Application No. 19723988.2.
Final Office Action dated Sep. 24, 2021 in U.S. Appl. No. 16/788,743.
Final Office Action dated Oct. 1, 2021 in U.S. Appl. No. 16/677,012.
Final Office Action dated Nov. 2, 2021 in U.S. Appl. No. 16/789,311.
Final Office Action dated Jan. 18, 2022 in U.S. Appl. No. 16/588,405.
Gertz et al., “Transposase mediated construction of RNA-seq libraries,” Genome Research 2012, 22, 134-141.
International Search Report and Written Opinion dated Sep. 22, 2021, in PCT Application No. PCT/US2021/013747.
International Search Report and Written Opinion dated Sep. 27, 2021, in PCT Application No. PCT/US2021/017719.
International Search Report and Written Opinion dated Oct. 12, 2021, in PCT Application No. PCT/US2021/041327.
International Search Report and Written Opinion dated Oct. 29, 2021, in PCT Application No. PCT/US2021/032319.
International Search Report and Written Opinion dated Nov. 12, 2021, in PCT Application No. PCT/US2021/044036.
Lan et al., “Droplet barcoding for massively parallel single-molecule deep sequencing,” Nature Communications 2016, 7(11784), in 10 pages.
Mair et al., “A Targeted Multi-omic Analysis Approach Measures Protein Expression and Low-Abundance Transcripts on the Single-Cell Level”, Cell Reports 2020, 31(1), 107499, in 20 pages.
Non-Final Office Action dated Sep. 28, 2021 in U.S. Appl. No. 16/400,885.
Non-Final Office Action dated Sep. 30, 2021 in U.S. Appl. No. 16/374,626.
Non-Final Office Action dated Oct. 1, 2021 in U.S. Appl. No. 16/677,012.
Non-Final Office Action dated Oct. 8, 2021 in U.S. Appl. No. 16/400,866.
Non-Final Office Action dated Dec. 21, 2021 in U.S. Appl. No. 15/055,407.
Non-Final Office Action dated Jan. 6, 2022 in U.S. Appl. No. 15/084,307.
Notice of Allowance dated Nov. 16, 2021 in U.S. Appl. No. 16/836,750.
Office Action dated Aug. 30, 2021 in Japanese Patent Application No. 2019-540515.
Office Action dated Aug. 31, 2021, in Korean Patent Application No. 10-2019-7038794.
Office Action dated Sep. 14, 2021, in Chinese Patent Application No. 2016800523302.
Office Action dated Oct. 21, 2021, in Chinese Patent Application No. 2016800073512.
Office Action dated Dec. 23, 2021, in Japanese Patent Application No. 2019-566787.
Office Action dated Dec. 17, 2021 in Korean Patent Application No. 10-2018-7008560.
Quail et al., “SASI-Seq: sample assurance Spike-Ins, and highly differentiating 384 barcoding for Illumina sequencing,” BMC Genomics 2014, 15(110), in 13 pages.
Restriction Requirement dated Oct. 1, 2021 in U.S. Appl. No. 16/232,287.
Restriction Requirement dated Dec. 27, 2021 in U.S. Appl. No. 16/747,737.
Schouten et al., “Relative quantification of 40 nucleic acid sequences by multiplex ligation-dependent probe amplification,” Nucleic Acids Research 2002, 30(12), e57.
Shapiro et al., “Single-cell sequencing-based technologies will revolutionize whole-organism science,” Nature Reviews Genetics 2013, 14, 618-629.
Song et al., DNase-seq: a high-resolution technique for mapping active gene regulatory elements across the genome from mammalian cells, Cold Spring Harb Protoc 2010, 2, in 13 pages.
Sos et al., “Characterization of chromatin accessibility with a transposome hypersensitive sites sequencing (THS-seq) assay,” Genome Biology 2016, 17(20), in 15 pages.
Trzupek et al., “Discovery of CD80 and CD86 as recent activation markers on regulatory T cells by protein-RNA single-cell analysis”, Genome Medicine 2020, 12(1), in 22 pages.
Wang et al., “Tagmentation-based whole-genome bisulfite sequencing,” Nature Protocols 2013, 8(10), 2022-2032.
Zhao et al., “Methylated DNA Immunoprecipitation and High-Throughput Sequencing (MeDIP-seq) Using Low Amounts of Genomic DNA,” Cellular Reprogramming 2014, 16(3), in 20 pages.
Related Publications (1)
Number Date Country
20180216174 A1 Aug 2018 US
Provisional Applications (1)
Number Date Country
62453163 Feb 2017 US