Assessing graft suitability for transplantation

Information

  • Patent Grant
  • 12084720
  • Patent Number
    12,084,720
  • Date Filed
    Friday, December 14, 2018
    6 years ago
  • Date Issued
    Tuesday, September 10, 2024
    3 months ago
Abstract
This invention relates to methods and compositions for assessing the suitability of a graft for transplantation or implantation by measuring total and/or specific cell-free nucleic acids (such as cf-DNA) and/or cell lysis. Specifically, the method comprising obtaining an amount of total cf DNA and/or graft-specific cfDNA released from a potential graft (e.g., ex vivo), e.g., prior to contacting of the potential graft with blood cells of a potential recipient, and/or subsequent to contacting of the potential graft or cells thereof with blood cells from a potential recipient, assessing the amount(s) to determine the suitability of the potential graft for transplantation or implantation.
Description
FIELD OF THE INVENTION

This invention relates to methods and related compositions for assessing the suitability of a graft for transplantation or implantation by measuring total and/or graft-specific cell-free nucleic acids, such as cell-free DNA.


SUMMARY OF INVENTION

In one aspect, a method of assessing the suitability of a graft is provided.


In one embodiment of any one of the methods provided herein, the method further comprises obtaining the one or more samples.


In one embodiment of any one of the methods provided herein, the value for the amount of total cell-free nucleic acids (such as DNA) and/or value for the amount of specific cell-free nucleic acids (such as DNA) are provided in a report. In one aspect, a report with one or more of the values obtained by any one of the methods provided herein is provided.


In one embodiment, any one of the methods provided can further comprise obtaining a value for the amount of total cell-free nucleic acids (such as DNA) in one or more other samples, and/or obtaining a value for the amount of specific cell-free nucleic acids (such as DNA) in one or more other samples, wherein the one or more other samples are from a subsequent time point or points.


In one embodiment of any one of the methods provided herein, the one or more samples and/or one or more other samples are obtained within minutes, such as no more than 15, 20, 25, 30, 35, 40, 45, 50, or 55 minutes, of obtaining the graft (e.g., storing the graft, perfusing the graft, etc.).


In one embodiment of any one of the methods provided herein, the one or more samples and/or one or more other samples are obtained within hours, such as no more than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 18 or more hours, of obtaining the graft (e.g., storing the graft, perfusing the graft, etc.).


In one embodiment of any one of the methods provided herein, an initial sample is obtained within an hour of obtaining the graft and one or more other samples are obtained within 15, 20, 25, 30, 35, 40, 45, 50, or 55 minute intervals or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 18 or more hourly intervals, such as until a threshold value or baseline is reached.


In one embodiment of any one of the methods provided herein, the one or more other subsequent time points are at hourly intervals. In one embodiment of any one of the methods provided herein, the one or more other subsequent time points are at daily intervals. In one embodiment of any one of the methods provided herein, the one or more other subsequent time points are at one-week intervals. In one embodiment of any one of the methods provided herein, the one or more other subsequent time points are at two-week intervals. In one embodiment of any one of the methods provided herein, the one or more other subsequent time points are at monthly intervals.


In one embodiment of any one of the methods provided herein, the specific cell-free nucleic acids (such as DNA) are graft-specific cell-free nucleic acids (such as DNA).


In one embodiment of any one of the methods provided herein, the method further comprises obtaining the one or more samples and/or one or more other samples. In one embodiment of any one of the methods provided herein, the method further comprises providing the one or more samples.


In one embodiment of any one of the methods provided herein, the monitoring of the graft comprises any one of the methods provided herein.


In one embodiment of any one of the methods provided herein, the sample comprises media, blood, plasma or serum.


In one aspect, a report comprising any one or more of the values provided herein is provided. In one embodiment of any one of the reports provided, the report comprises a value for the amount of total cell-free nucleic acids (such as DNA) in one or more samples and/or a value for the amount of specific cell-free nucleic acids (such as DNA) in one or more samples.


In one embodiment of any one of the reports provided, the report further comprises a value for the amount of total cell-free nucleic acids (such as DNA) from one or more other samples and/or a value for the amount of specific cell-free nucleic acids (such as DNA) from one or more other samples, wherein the one or more other samples are from a subsequent time point or points. In one embodiment of any one of the reports provided, the subsequent time point is at least one day later. In one embodiment of any one of the reports provided, the subsequent time point is at least one week later. In one embodiment of any one of the reports provided, the subsequent time point is at least two weeks later. In one embodiment of any one of the reports provided, the subsequent time point is at least a month later.


In one embodiment of any one of the methods provided herein, the method for obtaining an amount of total cell-free nucleic acids (such as DNA) comprises amplification, such as with real-time PCR or digital PCR. In one embodiment of any one of such methods comprising amplification, such as with real-time PCR or digital PCR, one or more targets are amplified. In one embodiment of any one of these methods, RNase P is the target or one of the targets for amplification. Any of a number of reference genes can be amplified for the analysis. Other reference genes that can serve as the target for amplification will be known to those of ordinary skill in the art.


In one embodiment of any one of such methods provided herein, the methods for obtaining an amount of specific cell-free nucleic acids (such as DNA) (for example, when a graft is a xenograft) comprises amplification, such as with real-time PCR. In one embodiment of any one of such methods, the method comprises, obtaining a quantification of one or more targets specific to the graft and one or more targets specific to the recipient or potential recipient. In one embodiment of any one of the methods provided herein, the method further comprises obtaining the one or more graft-specific targets and/or the one or more recipient or potential recipient targets. In one embodiment of any one of the methods provided herein, the quantification is obtained for each target relative to a standard, such as an internal standard, that may be spiked into a sample(s).


In one embodiment of any one of such methods provided herein, the methods for obtaining an amount of specific cell-free nucleic acids (such as DNA) can comprise a mismatch PCR amplification method. In one embodiment of any one of the methods provided herein, such a mismatch method comprises, for each of a plurality of single nucleotide variant (SNV) targets, obtaining results from an amplification-based quantification assay, such as a polymerase chain reaction (PCR) quantification assay, on a sample, or portion thereof, with at least one primer pair, wherein the at least one primer pair comprises a forward primer and a reverse primer, wherein the at least one primer pair comprises a primer with a 3′ mismatch (e.g., penultimate mismatch) relative to one sequence (e.g., allele) of the SNV target but a 3′ double mismatch relative to another sequence (e.g., allele) of the SNV target and specifically amplifies the one sequence (e.g., allele) of the SNV target.


In one embodiment of any one of the methods provided herein, such a mismatch method further comprises, for each SNV target, obtaining results from a quantification assay with at least one another primer pair, wherein the at least one another primer pair comprises a forward primer and a reverse primer, wherein the at least one another primer pair specifically amplifies another sequence (e.g., allele) of the SNV target.


In one embodiment of any one of the methods provided herein, such a mismatch method comprises, for each of a plurality of single nucleotide variant (SNV) targets, performing an amplification-based quantification assay, such as a PCR quantification assay, on a sample, or portion thereof, with at least two primer pairs, wherein each primer pair comprises a forward primer and a reverse primer, wherein one of the at least two primer pairs comprises a 3′ mismatch (e.g., penultimate) relative to one sequence (e.g., allele) of the SNV target but a 3′ double mismatch relative to another sequence (e.g., allele) of the SNV target and specifically amplifies the one sequence (e.g., allele) of the SNV target, and another of the at least two primer pairs specifically amplifies the another sequence (e.g., allele) of the SNV target.


In one embodiment of any one of the methods provided herein, such a mismatch method comprises obtaining results from an amplification-based amplification assay, such as a polymerase chain reaction (PCR) quantification assay, for each of a plurality of single nucleotide variant (SNV) targets, performed on a sample, or portion thereof, with at least two primer pairs, wherein each primer pair comprises a forward primer and a reverse primer, wherein one of the at least two primer pairs comprises a 3′ mismatch (e.g., penultimate) relative to one sequence (e.g., allele) of the SNV target but a 3′ double mismatch relative to another sequence (e.g., allele) of the SNV target and specifically amplifies the one sequence (e.g., allele) of the SNV target, and another of the at least two primer pairs specifically amplifies the another sequence (e.g., allele) of the SNV target.


In one embodiment of any one of the methods provided herein, such a mismatch method comprises obtaining results from an amplification-based quantification assay, such as a polymerase chain reaction (PCR) assay on a sample with at least one primer pair as provided herein, such as at least two primer pairs, wherein each primer pair comprises a forward primer and a reverse primer, selecting informative results based on the genotype of the specific nucleic acids and/or non-specific nucleic acids, and determining the amount of the non-specific nucleic acids in the sample based on the informative results. In one embodiment of any one of the methods provided herein, such a mismatch method further comprises identifying the plurality of SNV targets. In one embodiment of any one of the methods provided herein, such a mismatch method further comprises inferring the genotype of the non-specific nucleic acids.


In one embodiment of any one of the methods provided herein, such a mismatch method comprises obtaining results from 1) an amplification-based quantification assay, such as a PCR quantification assay, for each of a plurality of SNV targets, performed on a sample, or portion thereof, with at least one primer pair, such as at least two primer pairs, wherein each primer pair comprises a forward primer and a reverse primer, wherein one of the at least one, such as at least two, primer pair, comprises a 3′ mismatch (e.g., penultimate) relative to one sequence (e.g., allele) of the SNV target but a 3′ double mismatch relative to another sequence (e.g., allele) of the SNV target and specifically amplifies the one sequence (e.g., allele) of the SNV target and 2) a determination of informative results based on the specific genotype and/or a prediction of the likely non-specific genotype. In one embodiment of any one of such mismatch methods, when there are at least two primer pairs, the another primer pair specifically amplifies the another sequence (e.g., allele) of each SNV target and quantification results are obtained with the another primer pair for each of the SNV targets.


In one embodiment of any one of the methods provided herein, such a mismatch method comprises obtaining results from 1) an amplification-based quantification assay, such as a PCR quantification assay, for each of a plurality of SNV targets, performed on a sample, or portion thereof, with at least two primer pairs, wherein each primer pair comprises a forward primer and a reverse primer, wherein one of the at least two primer pairs comprises a 3′ mismatch (e.g., penultimate) relative to one sequence (e.g., allele) of the SNV target but a 3′ double mismatch relative to another sequence (e.g., allele) of the SNV target and specifically amplifies the one sequence (e.g., allele) of the SNV target, and another of the at least two primer pairs specifically amplifies the another sequence (e.g., allele) of the SNV target, and 2) a determination of informative results based on the specific genotype and/or a prediction of the likely non-specific genotype.


In one embodiment of any one of the methods provided herein, such a mismatch method further comprises at least one another primer pair for each SNV target and/or obtaining results with an amplification-based quantification assay, such as a PCR quantification assay therewith. In one embodiment of any one of such mismatch methods, the at least one another primer pair comprises a 3′ mismatch (e.g., penultimate) relative to another sequence (e.g., allele) of the SNV target but a 3′ double mismatch relative to the one sequence (e.g., allele) of the SNV target and specifically amplifies the another sequence (e.g., allele) of the SNV target.


In one embodiment of any one of the methods provided herein, such a mismatch method further comprises assessing the amount of specific nucleic acids based on the results.


In one embodiment of any one of such mismatch methods, the results are informative results.


In one embodiment of any one of such mismatch methods, the method further comprises selecting informative results of the amplification-based quantification assays, such as PCR quantification assays. In one embodiment of any one of such mismatch methods, the selected informative results are averaged, such as a median average. In one embodiment of any one of such mismatch methods, the results can be further analyzed with Robust Statistics. In one embodiment of any one of such mismatch methods, the results can be further analyzed with a Standard Deviation, such as a Robust Standard Deviation, and/or Coefficient of Variation, such as a Robust Coefficient of Variation, or % Coefficient of Variation, such as a % Robust Coefficient of Variation.


In one embodiment of any one of such mismatch methods, the informative results of the amplification-based quantification assays, such as PCR quantification assays are selected based on the genotype of the non-specific nucleic acids and/or specific nucleic acids.


In one embodiment of any one of such mismatch methods, the method further comprises obtaining the genotype of the non-specific nucleic acids and/or specific nucleic acids.


In one embodiment of any one of such mismatch methods, there is at least one primer pair, at least two primer pairs, at least three primer pairs, at least four primer pairs or more per SNV target. In one embodiment of any one of such mismatch methods, the plurality of SNV targets is at least 45, 48, 50, 55, 60, 65, 70, 75, 80, 85 or 90 or more. In one embodiment of any one of such mismatch methods, the plurality of SNV targets is at least 90, 95 or more targets. In one embodiment of any one of such mismatch methods, the plurality of SNV targets is less than 90, 95 or more targets. In one embodiment of any one of such mismatch methods, the plurality of SNV targets is less than 105 or 100 targets.


In one embodiment of any one of such mismatch methods, the mismatched primer(s) is/are the forward primer(s). In one embodiment of any one of such mismatch methods, the reverse primers for the primer pairs for each SNV target is the same.


In one embodiment of any one of the methods provided herein, the amount of the specific cell-free nucleic acids (such as DNA) is the ratio or percentage of specific nucleic acids to total or non-specific nucleic acids.


In one embodiment of any one of the methods provided herein, the method further comprises extracting nucleic acids from the sample.


In one embodiment of any one of the methods provided herein, the method further comprises a pre-amplification step. In one embodiment of any one of the methods provided herein, the pre-amplification is performed prior to the quantification assay(s).


In one embodiment, any one of the embodiments for the methods provided herein can be an embodiment for any one of the reports provided. In one embodiment, any one of the embodiments for the reports provided herein can be an embodiment for any one of the methods provided herein.





BRIEF DESCRIPTION OF FIGURES

The accompanying figures are not intended to be drawn to scale. The figures are illustrative only and are not required for enablement of the disclosure.



FIG. 1 provides an exemplary, non-limiting diagram of MOMA primers. In a polymerase chain reaction (PCR) assay, extension of the sequence containing SNV A is expected to occur, resulting in the detection of SNV A, which may be subsequently quantified. Extension of the SNV B, however, is not expected to occur due to the double mismatch.



FIG. 2 shows exemplary outline for determining cell-free nucleic acids (such as DNA).



FIG. 3 is a workflow schematic depicting one embodiment of a sample processing scheme.



FIG. 4 shows the proportion of long fragment DNA in the eluates (reported as the mean of duplicate determinations) in the control (normal plasma specimen) and tubes 2-5.



FIG. 5 is a representative electropherogram image of a fragment distribution in Tubes 2-5. The peak height indicates the fragment levels. The fragment length (BP) is indicated on the trace. FU=fluorescence units.



FIG. 6 shows the STEEN/plasma supernatant after 1400×g spin, showing essential absence of leukocytes, similar to that seen in plasma collections in the absence of STEEN (magnification 400×) (left panel). For comparison, the resuspended 200 μL 1400×g pellet (magnification 400×) showing effective removal of any residual leukocytes in the post-PPT STEEN/plasma supernatant by the subsequent 1400×g spin step, congruent with results seen using plasma without STEEN (right panel).



FIG. 7 is a graph showing the percent recovery of different concentrations of gDNA and heparin added to STEEN™ solution.





DETAILED DESCRIPTION OF THE INVENTION

Total cell free DNA of a graft ex vivo in a perfusion container generally will represent lysis and/or apoptosis of cells from the graft and any cells from blood from the donor. Without being bound by theory, it is thought that as a graft starts to deteriorate, apoptosis of cells increase, and total cell free DNA levels will also increase. A suitable graft for transplant generally has low or steady-state levels of total cell free DNA.


Aspects of the disclosure relate to methods for assessing the suitability of a graft. Methods provided herein or otherwise known in the art can be used multiple times to obtain total and/or specific cell-free nucleic acid (such as DNA) values over time. Also included are reports that can include one or more of these values. Such reports can provide valuable information to a clinician. In some embodiments, the clinician can then assess the condition (or suitability of a graft) and/or make treatment decisions accordingly for a subject.


As used herein, “graft” refers to a biological material comprising cells or tissue, such as at least a portion of an organ, that may be transplanted or implanted in or into a subject. In some embodiments, the graft is explanted material comprising cells or tissue, such as at least a portion of an organ that is being maintained outside the body (ex vivo), such as to preserve or rehabilitate, the graft. Any one of the methods provided herein can be used to evaluate its suitability for future engraftment. In one embodiment of any one of the methods provided herein, the material are EVLP lungs, such as after removal from a subject and before engraftment into another subject.


In some embodiments of any one of the methods provided herein, the graft is a whole organ or more than one organ. Examples of organs that can be transplanted or implanted include, but are not limited to, the heart, kidney(s), kidney, liver, lung(s), pancreas, intestine, etc. An example of more than one organ includes a combination of a heart and lung. In other embodiments of any one of the methods provided herein, the graft is less than a whole organ and is at most a portion thereof, such as a valve. Grafts may be of the same species or may be of a different species.


Accordingly, in some embodiments of any one of the methods provided herein, the graft is from a different species (or is a xenograft), such as from a pig or cow when the recipient is other than a pig or cow, respectively, such as a human. Any one of the types of grafts provided herein may be a xenograft. In some embodiments of any one of the methods provided herein, the graft is a pig or cow valve. In other embodiments of any one of the methods provided herein, the graft is from the same species. In other embodiments of any one of the methods provided herein the graft is decellularized graft, such as a decellularized xenograft. In some embodiments of any one of the methods provided herein the graft is an autograft. Any one of the methods or compositions provided herein may be used for assessing any one of the grafts described herein.


As used herein, the sample can be a biological sample. Examples of such biological samples include whole blood, plasma, serum, etc.


In one embodiment of any one of the methods provided herein, the sample may be of or comprise media in which the graft is placed or with which it has contact. In one embodiment of any one of such samples, the media can comprise blood or a blood substitute, preservation solution, or any other solution in which a graft can be placed or with which it has contact, such as in in vitro contexts. In one embodiment of any one of such samples the graft, such as an organ or organs can be contained in a perfusion system.


In one embodiment of any one of the methods provided herein, the graft (e.g., cells, tissue, organ) is maintained in graft storage media. Graft storage media, such as organ preservation solutions, are well known in the art. Graft storage media can be intracellular (e.g., perfused) or extracellular, and may depend on the graft to be preserved. Approaches to preserving most grafts include simple static cold storage (SCS) and dynamic preservation. Examples of dynamic preservation include hypothermic machine perfusion (HMP), normothermic machine perfusion, and oxygen persufflation. Typically, in combination with hypothermia, graft storage media can prevent clotting in harvests with blood present, reduce stress and deterioration associated with ex vivo handling, and decrease the risk of microbial growth. Therefore, in some embodiments, the graft storage media can comprise osmotic active agents, electrolytes, hydrogen ion buffers, colloid(s), metabolic inhibitors, metabolites, and antioxidants. Examples of osmotic active agents, which may prevent cell swelling, include lactobionate, raffinose, citrate, and gluconate. Electrolytes, which can exert an osmotic effect, include sodium, potassium, calcium, and magnesium ions. Examples of hydrogen ion buffers include phosphate, histidine, and N-(2-hydroxyethyl)-piperazine-N′-2-ethanesulfonic acid (HEPES) buffer. Examples of colloids, which may be used during the initial vascular flush out and perfusion, include albumin and HES. Examples of metabolic inhibitors, which may suppress degradation of cell constituents, include allopurinol, antiproteases, and chlorpromazine. Examples of metabolites, which can help restore metabolism during the reperfusion phase, include adenosine, glutathione, and phosphate. Examples of antioxidants, which can inhibit oxygen free-radical injury, include steroids, vitamin E, deferoxamine, and tryptophan.


Graft storage media are commercially available, and examples include BELZER UW® cold storage solution (VIASPAN™ or the University of Wisconsin (UW) solution), CELSIOR®, CUSTODIOL®, and IGL-1®.


In some aspects, the methods include steps for determining a value for the amount of total cell-free nucleic acids (such as DNA) and/or a value for the amount of specific cell-free nucleic acids (such as DNA).


As used herein, a “value” is any indicator that conveys information about an “amount”. The indicator can be an absolute or relative value for the amount. As used herein, “amount” refers to the quantity of nucleic acids (such as DNA). Further, the value can be the amount, frequency, ratio, percentage, etc.


In some instances the values can be compared to a “threshold value”. As used herein, a “threshold value” refers to any predetermined level or range of levels that is indicative of a state, the presence or absence of a condition or the presence or absence of a risk. The threshold value can take a variety of forms. It can be single cut-off value, such as a median or mean. It can be established based upon comparative groups, such as where the risk in one defined group is double the risk in another defined group. As another example, a threshold value is a baseline value, such as without the presence of a state, condition or risk or after a course of treatment or other remedial action. Such a baseline can be indicative of a normal or other state not correlated with the risk or condition or state that is being tested for.


As used herein, “specific cell-free nucleic acids” refers to a subset of cell-free nucleic acids (such as DNA) that is within total cell-free nucleic acids (such as DNA). In some embodiments, the specific cell-free nucleic acids (such as DNA) are cell-free nucleic acids (such as DNA) that are graft-specific (GS). GS cf-DNA refers to DNA that presumably is shed from the graft or cells thereof, the sequence of which matches (in whole or in part) the genotype of the subject from which the graft is obtained. As used herein, GS cf-DNA may refer to certain sequence(s) in the GS cf-DNA population, where the sequence is distinguishable from the recipient or potential recipient cf-DNA (e.g., having a different sequence at a particular nucleotide location(s)), or it may refer to the entire GS cf-DNA population).


The values for the amount(s) of nucleic acids (such as DNA) can be “obtained” by any one of the methods provided herein, and any obtaining step(s) can include any one of the methods incorporated herein by reference or otherwise provided herein. “Obtaining” as used herein refers to any method by which the respective information or materials can be acquired. Thus, the respective information can be acquired by experimental methods. Respective materials can be created, designed, etc. with various experimental or laboratory methods, in some embodiments. The respective information or materials can also be acquired by being given or provided with the information, such as in a report, or materials. Materials may be given or provided through commercial means (i.e. by purchasing), in some embodiments.


As provided herein, the suitability can be determined using one or more values for the amount of total cell-free nucleic acids (such as DNA) and/or one or more values for the amount of specific cell-free nucleic acids (such as DNA).


Ideally, most of the cell free DNA to be analyzed will come from the organ, and the blood will have washed away. However, intact leukocytes from the donor can still be present in the organ. Also, lysis of cells can lower the quality of the perfusate for total cell free DNA analysis. Thus, in some embodiments of any one of the methods provided herein, contaminating intact cells are removed from samples, such as perfusate samples, by one or more (e.g., one, two or three or more) centrifugation steps. In one embodiment of any one of the methods provided herein, a baseline can be established for meaningful cfDNA analysis after effective washout of contaminating leukocytes.


The suitability can also be determined using one or more values for the amount of total cell-free nucleic acids (such as DNA) and/or one or more values from fragment analysis.


Fragment analysis can be performed by assessing short and/or long nucleic acid fragments. As used herein, a “long fragment” refers to a fragment that is greater than 170 bps (e.g., between 171 and 300 bps in length), while a “short fragment” is a fragment that is less than or equal to 170 bps (e.g., between 75 and 170 bps in length). Such methods generally are performed with primers targeting a long fragment and/or a short fragment.


The fragment can be an Alu fragment. An Alu element is a short stretch of DNA originally characterized by the action of the Arthrobacter luteus (Alu) restriction endonuclease. Alu repeats are the most abundant sequences in the human genome, with a copy number of about 1.4 million per genome. Alu sequences are short interspersed nucleotide elements (SINEs), typically 300 nucleotides, which account for more than 10% of the genome. Provided herein are methods that in one embodiment can include measuring the potential contaminating contribution of cell lysis of a cf-DNA sample by analyzing long Alu fragments and/or short Alu fragments.


In some embodiments of any one of the methods provided, the method further includes assessing the suitability (e.g., health, state, or condition) of a graft for transplantation or implantation based on the value(s). In some embodiments, any one of the methods provided herein can comprise correlating an increase in one or more values (e.g., for an amount of total and/or specific cell-free nucleic acids (such as DNA)) with unsuitability or declining suitability or a decrease in one or more values (e.g., for an amount of total and/or specific cell-free nucleic acids (such as DNA)) with suitability or increasing suitability. In some embodiments of any one of the methods provided herein, correlating comprises comparing a level (e.g., concentration, ratio or percentage) to a threshold value or value from another point in time to determine suitability, or increasing or decreasing suitability. Thus, changes in the levels can be monitored over time. Any one of the methods provided herein can include one or more steps of comparing the values for an amount of nucleic acids (such as DNA) to a threshold value or a value from a different point in time to assess the suitability of the graph.


In one embodiment of any one of the methods provided herein, the method may further includes an additional test(s) for assessing. The additional test(s) may be any one of the methods provided herein or methods known in the art.


It has been found that particularly useful to a clinician is a report that contains the value(s) provided herein. In some embodiments of any one of the reports provided, the reports also include one or more threshold values. In one aspect, therefore such reports are provided. Reports may be in oral, written (or hard copy) or electronic form, such as in a form that can be visualized or displayed. In some embodiments, the “raw” results for each assay as provided herein are provided in a report, and from this report, further steps can be taken to analyze the amount(s) nucleic acids (such as DNA). In other embodiments, the report provides multiple values for the amounts of nucleic acids (such as DNA). From the amounts, in some embodiments, a clinician may assess the suitability of a graft for transplantation or implantation or the need to monitor the graft over time or treatment or some other remedial action.


In some embodiments, the amounts are in or entered into a database. In one aspect, a database with such values is provided. From the amount(s), a clinician may assess the need for a treatment or monitoring. Accordingly, in any one of the methods provided herein, the method can include assessing the amount(s) at more than one point in time. Such assessing can be performed with any one of the methods or compositions provided herein.


In any one of the methods provided herein, the method can include assessing the amount of nucleic acids (such as DNA) at another point in time or times. Such assessing can be performed with any one of the methods provided herein.


Methods for determining total cell-free nucleic acids (such as DNA) as well as specific cell-free nucleic acids (such as DNA) are provided herein or are otherwise known in the art. For example, the methods of PCT Application No. PCT/US2016/030313 may be used for determining a value for the amount of specific cell-free nucleic acids (such as DNA) in a sample as provided herein. Thus, any one of the methods provided herein may include the steps of any one of the methods described in PCT Application No. PCT/US2016/030313, and such methods and steps are incorporated herein by reference. Likewise, the methods of measuring cell-free DNA of U.S. Publication No. US-2015-0086477-A1 are also incorporated herein by reference and such methods can be included as part of any one of the methods provided herein for determining a value for the amount of cell-free nucleic acids (such as DNA).


As a further example, amplification with PCR, such as real-time PCR or digital PCR, may be used to determine a value for the amount of total cell-free nucleic acids (such as DNA) and/or specific cell-free nucleic acids (such as DNA). For example, in some embodiments of any one of the methods provided herein, the total cell-free nucleic acids (such as DNA) is determined with Taqman Real-time PCR using RNase P as a target. Other methods are provided elsewhere herein or would be apparent to those of ordinary skill in the art. Any one of the methods provided herein, can include any one of the methods of determining a value provided herein.


As mentioned above, in some embodiments, any one of the methods provided herein may include steps of a quantitative assay that makes use of mismatch amplification (e.g., MOMA) in order to determine a value for an amount of specific cell-free nucleic acids (such as DNA). Primers for use in such assays may be obtained, and any one of the methods provided herein can include a step of obtaining one or more primer pairs for performing the quantitative assays. Generally, the primers possess unique properties that facilitate their use in quantifying amounts of nucleic acids. For example, a forward primer of a primer pair can be mismatched at a 3′ nucleotide (e.g., penultimate 3′ nucleotide). In some embodiments of any one of the methods provided, this mismatch is at a 3′ nucleotide but adjacent to the SNV position. In some embodiments of any one of the methods provided, the mismatch positioning of the primer relative to a SNV position is as shown in FIG. 1. Generally, such a forward primer even with the 3′ mismatch to produce an amplification product (in conjunction with a suitable reverse primer) in an amplification reaction, thus allowing for the amplification and resulting detection of a nucleic acid with the respective SNV. If the particular SNV is not present, and there is a double mismatch with respect to the other allele of the SNV target, an amplification product will generally not be produced. Preferably, in some embodiments of any one of the methods provided herein, for each SNV target a primer pair is obtained whereby specific amplification of each allele can occur without amplification of the other allele(s).


“Specific amplification” refers to the amplification of a specific target without substantial amplification of another nucleic acid or without amplification of another nucleic acid sequence above background or noise. In some embodiments, specific amplification results only in the amplification of the specific allele.


As used herein, “single nucleotide variant” refers to a nucleic acid sequence within which there is sequence variability at a single nucleotide. In some embodiments, the SNV is a biallelic SNV, meaning that there is one major allele and one minor allele for the SNV. In some embodiments, the SNV may have more than two alleles, such as within a population. Generally, a “minor allele” refers to an allele that is less frequent in a set of nucleic acids, for a locus, while a “major allele” refers to the more frequent allele in a set of nucleic acids. The methods provided herein can quantify nucleic acids of major and minor alleles within a mixture of nucleic acids even when present at low levels, in some embodiments.


The nucleic acid sequence within which there is sequence identity variability, such as a SNV, is generally referred to as a “target”. As used herein, a “SNV target” refers to a nucleic acid sequence within which there is sequence variability at a single nucleotide. The SNV target has more than one allele, and in preferred embodiments, the SNV target is biallelic. In some embodiments of any one of the methods provided herein, the SNV target is a SNP target. In some of these embodiments, the SNP target is biallelic. In some embodiments of any one of the methods provided, the amount of nucleic acids is determined by attempting amplification-based quantitative assays, such as quantitative PCR assays, with primers for a plurality of SNV targets. A “plurality of SNV targets” refers to more than one SNV target where for each target there are at least two alleles. Preferably, in some embodiments, each SNV target is expected to be biallelic and a primer pair specific to each allele of the SNV target is used to specifically amplify nucleic acids of each allele, where amplification occurs if the nucleic acid of the specific allele is present in the sample.


In some embodiments of any one of the methods provided herein, for each SNV target that is biallelic, there are two primer pairs, each specific to one of the two alleles and thus have a single mismatch with respect to the allele it is to amplify and a double mismatch with respect to the allele it is not to amplify (again if nucleic acids of these alleles are present). In some embodiments of any one of the methods provided herein, the mismatch primer is the forward primer. In some embodiments of any one of the methods provided herein, the reverse primer of the two primer pairs for each SNV target is the same.


These concepts can be used in the design of primer pairs for any one of the methods provided herein. It should be appreciated that the forward and reverse primers are designed to bind opposite strands (e.g., a sense strand and an antisense strand) in order to amplify a fragment of a specific locus of the template. The forward and reverse primers of a primer pair may be designed to amplify a nucleic acid fragment of any suitable size to detect the presence of, for example, an allele of a SNV target according to the disclosure. Any one of the methods provided herein can include one or more steps for obtaining one or more primer pairs as described herein.


Generally, “informative results” as provided herein are the results that can be used to quantify the level of nucleic acids in a sample. In some embodiments of any one of the methods provided, the amount of specific- and/or non-specific nucleic acids represents an average across informative results for the nucleic acids, respectively. In some embodiments of any one of the methods provided herein, this average is given as an absolute amount or as a percentage. Preferably, in some embodiments of any one of the methods provided herein, this average is the median.


The amount, such as ratio or percentage, of specific nucleic acids may be determined with the quantities of the major and minor alleles as well as genotype, as needed. In some embodiments of any one of the methods provided herein, the alleles can be determined based on prior genotyping (e.g., of the recipient or potential recipient and/or the subject from which a graft is obtained, respectively). Methods for genotyping are well known in the art. Such methods include sequencing, such as next generation, hybridization, microarray, other separation technologies or PCR assays. Any one of the methods provided herein can include steps of obtaining such genotypes.


It should be appreciated that the primer pairs described herein may be used in a multiplex assays, such as multiplex PCR assays. Accordingly, in some embodiments, the primer pairs are designed to be compatible with other primer pairs in a PCR reaction. For example, the primer pairs may be designed to be compatible with at least 2, at least 5, at least 10, at least 20, at least 30, at least 40, etc. other primer pairs in a PCR reaction. As used herein, primer pairs in a PCR reaction are “compatible” if they are capable of amplifying their target in the same PCR reaction. In some embodiments, primer pairs are compatible if the primer pairs are inhibited from amplifying their target nucleic acid (such as DNA) by no more than 1%, no more than 2%, no more than 5%, no more than 10%, no more than 15%, no more than 20%, no more than 25%, no more than 30%, no more than 35%, no more than 40%, no more than 45%, no more than 50%, or no more than 60% when multiplexed in the same PCR reaction. Primer pairs may not be compatible for a number of reasons including, but not limited to, the formation of primer dimers and binding to off-target sites on a template that may interfere with another primer pair. Accordingly, the primer pairs of the disclosure may be designed to prevent the formation of dimers with other primer pairs or limit the number of off-target binding sites. Exemplary methods for designing primers for use in a multiplex assays are known in the art and are otherwise described herein.


In some embodiments of any one of the methods provided herein, the mismatch amplification-based quantitative assay is any quantitative assay whereby nucleic acids are amplified and the amounts of the nucleic acids can be determined. Such assays include those whereby nucleic acids are amplified with the MOMA primers as described herein and quantified. Such assays include simple amplification and detection, hybridization techniques, separation technologies, such as electrophoresis, next generation sequencing and the like.


In some embodiments of any one of the methods provided herein, the quantitative assays include quantitative PCR assays. Quantitative PCR include real-time PCR, digital PCR, Taqman, etc. In some embodiments of any one of the methods provided herein the PCR is “Real-time PCR”. Such PCR refers to a PCR reaction where the reaction kinetics can be monitored in the liquid phase while the amplification process is still proceeding. In contrast to conventional PCR, real-time PCR offers the ability to simultaneously detect or quantify in an amplification reaction in real time. Based on the increase of the fluorescence intensity from a specific dye, the concentration of the target can be determined even before the amplification reaches its plateau.


In any one of the methods provided herein the PCR may be digital PCR. Digital PCR involves partitioning of diluted amplification products into a plurality of discrete test sites such that most of the discrete test sites comprise either zero or one amplification product. The amplification products are then analyzed to provide a representation of the frequency of the selected genomic regions of interest in a sample. Analysis of one amplification product per discrete test site results in a binary “yes-or-no” result for each discrete test site, allowing the selected genomic regions of interest to be quantified and the relative frequency of the selected genomic regions of interest in relation to one another be determined. In certain aspects, in addition to or as an alternative, multiple analyses may be performed using amplification products corresponding to genomic regions from predetermined regions. Results from the analysis of two or more predetermined regions can be used to quantify and determine the relative frequency of the number of amplification products. Using two or more predetermined regions to determine the frequency in a sample reduces a possibility of bias through, e.g., variations in amplification efficiency, which may not be readily apparent through a single detection assay. Methods for quantifying DNA using digital PCR are known in the art and have been previously described, for example in U.S. Patent Publication number US20140242582.


Any one of the methods provided herein can comprise extracting nucleic acids, such as cell-free DNA. Such extraction can be done using any method known in the art or as otherwise provided herein (see, e.g., Current Protocols in Molecular Biology, latest edition, or the QIAamp circulating nucleic acid kit or other appropriate commercially available kits). An exemplary method for isolating cell-free DNA from blood is described. Blood containing an anti-coagulant such as EDTA or DTA is collected. The plasma, which contains cf-DNA, is separated from cells present in the blood (e.g., by centrifugation or filtering). An optional secondary separation may be performed to remove any remaining cells from the plasma (e.g., a second centrifugation or filtering step). The cf-DNA can then be extracted using any method known in the art, e.g., using a commercial kit such as those produced by Qiagen. Other exemplary methods for extracting cf-DNA are also known in the art (see, e.g., Cell-Free Plasma DNA as a Predictor of Outcome in Severe Sepsis and Septic Shock. Clin. Chem. 2008, v. 54, p. 1000-1007; Prediction of MYCN Amplification in Neuroblastoma Using Serum DNA and Real-Time Quantitative Polymerase Chain Reaction. JCO 2005, v. 23, p. 5205-5210; Circulating Nucleic Acids in Blood of Healthy Male and Female Donors. Clin. Chem. 2005, v. 51, p. 1317-1319; Use of Magnetic Beads for Plasma Cell-free DNA Extraction: Toward Automation of Plasma DNA Analysis for Molecular Diagnostics. Clin. Chem. 2003, v. 49, p. 1953-1955; Chiu R W K, Poon L L M, Lau T K, Leung T N, Wong E M C, Lo Y M D. Effects of blood-processing protocols on fetal and total DNA quantification in maternal plasma. Clin Chem 2001; 47:1607-1613; and Swinkels et al. Effects of Blood-Processing Protocols on Cell-free DNA Quantification in Plasma. Clinical Chemistry, 2003, vol. 49, no. 3, 525-526).


In some embodiments of any one of the methods provided herein, a pre-amplification step is performed. An exemplary method of such a pre-amplification is as follows, and such a method can be included in any one of the methods provided herein. Approximately 15 ng of cell-free plasma DNA is amplified in a PCR using Q5 DNA polymerase with approximately 13 targets where pooled primers were at 4 uM total. Samples undergo approximately 25 cycles. Reactions are in 25 ul total. After amplification, samples can be cleaned up using several approaches including AMPURE bead cleanup, bead purification, or simply ExoSAP-IT™, or Zymo.


Various aspects of the present invention may be used alone, in combination, or in a variety of arrangements not specifically discussed in the embodiments described in the foregoing and are therefore not limited in their application to the details and arrangement of components set forth in the foregoing description or illustrated in the drawings. For example, aspects described in one embodiment may be combined in any manner with aspects described in other embodiments.


Also, embodiments of the invention may be implemented as one or more methods, of which an example has been provided. The acts performed as part of the method(s) may be ordered in any suitable way. Accordingly, embodiments may be constructed in which acts are performed in an order different from illustrated, which may include performing some acts simultaneously, even though shown as sequential acts in illustrative embodiments.


Use of ordinal terms such as “first,” “second,” “third,” etc., in the claims to modify a claim element does not by itself connote any priority, precedence, or order of one claim element over another or the temporal order in which acts of a method are performed. Such terms are used merely as labels to distinguish one claim element having a certain name from another element having a same name (but for use of the ordinal term).


The phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting. The use of “including,” “comprising,” “having,” “containing”, “involving”, and variations thereof, is meant to encompass the items listed thereafter and additional items.


Having described several embodiments of the invention in detail, various modifications and improvements will readily occur to those skilled in the art. Such modifications and improvements are intended to be within the spirit and scope of the invention. Accordingly, the foregoing description is by way of example only, and is not intended as limiting. The following description provides examples of the methods provided herein.


EXAMPLES
Example 1—Evaluation of Gel-Separator Plasma Preparation Tubes (PPT)

Eight mL of neat STEEN™ was added to a 8.5 mL PPT and spun for 10 minutes at 1100×g. For comparison, 1.0 mL of human buffycoat (a plasma-white blood cell mixture tinged with red blood cells) was added to 7.0 mL of neat STEEN™ and spun for 10 minutes at 1100×g in an 8.5 mL PPT tube. Physical observations included a finding of clarification after centrifugation of fluid-phase above the gel plug and migration below the plug of the cell fraction. The observed thin line at the top of the gel after separation was due to embedding to red blood cell fragments in the gel material, a phenomenon also seen when spinning whole blood into a plasma separator gel.


The experiment used a controlled buffy coat spike into STEEN solution to simulate a substantial leukocyte/RBC complement that may be present within the STEEN solution circulating through ex vivo lung perfusion (EVLP) lungs. Migration of cells through the gel separator to form a small red-tinged pellet at the bottom of the PPT below the gel separator indicates that cells suspended in STEEN™ solution pass through the gel plug of PPTs to form a pellet well-separated from the fluid phase in a manner analogous to that observed during centrifugation of human whole blood samples in a PPT. This indicates that, for samples of STEEN perfusate collecting during EVLP, the post-spin supernatant poured off a PPT will be suitable for cf-DNA analysis without problematic contamination by DNA from contaminating leukocytes.


As a result, a conditional paired control step demonstrating the cellular migration of buffy coat cells spiked into purified human plasma versus STEEN solution was performed. Plasma was purified for cf-DNA extraction and analysis by centrifuging a transplant recipient's whole blood in a PPT, then pouring off and briefly recentrifuging the PPT supernatant in a conical tube at low speed to pellet any residual cellular debris. In order to evaluate the degree of clearance of leukocytes at the microscopic level from the fluid phase of intra-EVLP STEEN perfusate (as opposed to whole blood), supernatant prepared as described above was poured off into a 15 mL conical tube and subjected to a second spin at 1400×g for 10 minutes. The supernatant from the second spin was then removed from the conical tube, leaving 200 μL at the bottom of the tube to prevent disruption of the small cellular pellet. The collected supernatant was examined microscopically by hemocytometry for any remaining cellularity. The 200 μL volume at the bottom of the tube including the pellet was resuspended and also viewed microscopically through a hemocytometer. FIG. 6 shows the microscopic results.


A single low speed spin of a buffycoat-STEEN™ mixture through a gel separator PPT, followed by a clean-up low speed (1400×g for 10 minutes) spin, leaves the remaining plasma-STEEN™ fluid phase essentially completely clear of contaminating leukocytes, allowing meaningful analysis of cf-DNA in STEEN™ perfusates. The experiment demonstrates that STEEN solution and its component molecules (Dextran 40, for example) does not substantially effect the ability of the protocol for plasma purification described above to similarly remove contaminating leukocytes from STEEN-based EVLP perfusion solutes collected for cf-DNA analysis.


Example 2—Determination of the Presence/Absence of Human DNA in Neat STEEN™

Four mL volumes of neat STEEN™ solution, a low cf-DNA positive extraction control (PEC; human plasma) and a negative extraction control (NEC; nuclease-free water) were extracted in triplicate using an automated DNA extraction workflow. Eluates from the extraction process were monitored for detection of a human reference gene by a highly sensitive PCR method validated for quantification of cf-DNA. For comparison, PCR detection assay was also applied to neat STEEN™ solution without extraction.









TABLE 1







Reference Gene Amplification









Evidence of Reference



Gene Amplification


Sample Type
(Yes/No)





STEEN ™-extracted (in triplicate)
No


STEEN ™-not extracted (in triplicate)
No


NEC-nuclease-free water-1
No


PEC-human plasma-1
Yes









No human DNA was detected in either the non-extracted STEEN™ or the extracted STEEN™. This indicates that baseline levels of human DNA in STEEN™ are at a minimum extremely low and below the level of detections for this highly sensitive assay. If STEEN™ does contain any human DNA, it is probably at a level too low to confound cf-DNA measurements.


Example 3—Evaluation of DNA in a Neat STEEN™ Solution Using Endogenous Long and Short Fragments of DNA

Three 4 mL extractions of neat STEEN™ solution were extracted and analyzed using the short and long fragment DNA tests in triplicate, including a positive extraction control (PEC; a human plasma sample), a negative extraction control (NEC; nuclease-free water), non-extracted STEEN™ solution, and non-extracted 0.1× TE buffer.


Long and short fragment cf-DNA was detected at levels historically expected in the normal, well-characterized PEC after standard automated cf-DNA extraction. In the same run, using this more sensitive long and short fragment quantification assay, and in agreement with the results obtained in Example 2 using the reference gene qPCR assay, no DNA amplification was detected in the NEC, the non-extracted STEEN™ solution, or the extracted STEEN™ solution. Thus, the STEEN™ solution was found to not contain human DNA at a level conceivably able to confound cf-DNA evaluations.


Example 4—Sheared Genomic DNA (gDNA) or Short Fragment gDNA Spike-In into STEEN™ Solution Versus Human Plasma (Quantitative Detection by Reference Gene qPCR)

Genomic DNA (gDNA) was fragmented in a controlled manner by sonication and spiked into a neat STEEN™ solution (without addition of additives, such as heparin) and plasma at defined concentrations, then extracted using an automated cf-DNA extraction methodology. Resulting concentrations of cf-DNA in the extraction eluates were measured using the reference gene qPCR method in order to determine the percent recovery of spiked-in DNA, as shown in Table 2.









TABLE 2







Percent Recovery of Sheared gDNA















Sheared
Mean




Sheared

gDNA
gDNA
gDNA



gDNA Input

Extraction
Extraction
Extraction


Sample #
(ng/mL)
Matrix
(ng/mL)
(ng/ml)
%















1
0
STEEN
0.0
0.0
 0%


2
0
STEEN
0.0




3
0.2
STEEN
0.2
0.1
66%


4
0.2
STEEN
0.1




5
1
STEEN
0.4
0.5
55%


6
1
STEEN
0.7




7
2.5
STEEN
0.8
0.9
36%


8
2.5
STEEN
1.0




9
5
STEEN
2.1
2.0
40%


10
5
STEEN
1.9




11
5
PLASMA
3.0
3.2
63%


12
5
PLASMA
3.3




13
15
STEEN
7.8
7.1
48%


14
15
STEEN
6.5




15
15
PLASMA
8.1
7.8
52%


16
15
PLASMA
7.5









Genomic DNA extraction efficiency is far from plasma or STEEN™ solution, congruent with known properties of extraction methodologies of multiple types, although the chemistry used for cf-DNA extraction was selected to be optimally efficient for efficient translation of short fragmented DNA (e.g., cf-DNA). Recognizing that normal plasma matrix has a low baseline content of cf-DNA, the data in Table 2 demonstrates that the automated cf-DNA methodology is capable of isolating total cf-DNA from human plasma.


To examine the same principles with short fragment gDNA, 25,000 copies of short DNA fragment control were spiked into samples of STEEN™ solution and human plasma containing a background of varying amounts of sheared human gDNA amounts, then subjected to an automated cf-DNA extraction protocol. The percent recovery of the short fragment in the extraction eluate was determined using the reference gene qPCR methods. The results are shown in Table 3 below.









TABLE 3







Percent Recovery of Short Fragment gDNA












Sheared

Short




gDNA

Fragment
Short



Input

Extraction
Fragment


Sample #
(ng/mL)
Matrix
(copies/mL)
Extraction %














1
0
STEEN
14448
58%


2
0
STEEN
9655
39%


3
0.2
STEEN
14868
59%


4
0.2
STEEN
14710
59%


5
1
STEEN
14310
57%


6
1
STEEN
11757
47%


7
2.5
STEEN
13961
56%


8
2.5
STEEN
15726
63%


9
5
STEEN
11627
47%


10
5
STEEN
12618
50%


11
5
PLASMA
16787
67%


12
5
PLASMA
16768
67%


13
15
STEEN
14723
59%


14
15
STEEN
14835
59%


15
15
PLASMA
17131
69%


16
15
PLASMA
14710
59%


17
PEC
PLASMA
0
 0%


18
NEC
NFW
0
 0%









Recognizing that normal plasma matrix has a low baseline content of cf-DNA, the data in Table 3 demonstrate that the automated cf-DNA extraction methodology is capable of isolating short DNA fragments from STEEN™ solutions with efficiency similar to that observed when extracting from human plasma. This is beneficial for the determination of cellular apoptosis from cellular lysis during the processing of EVLP samples.


Example 5—Evaluation of the Degree of PCR Inhibition by Heparin at Concentrations Used in the EVLP System

gDNA and heparin were added to STEEN™ solution at various concentrations up to 50 IU/mL. cf-DNA was extracted and quantified using an automated extraction system, and extraction efficiency was measuring using the reference gene qPCR. The results are shown in FIG. 7.


Inhibition of PCR, as measured by the quantification of a well-known reference gene is noticeable at approximately 50 IU/mL of heparin, a concentration of heparin typically used in whole blood unit donation bags. At this high concentration of heparin, recovery of the genomic reference gene drops dramatically, as shown in FIG. 7. Concentrations of heparin similar to blood collection tubes (approximately 15 IU/mL), concentrations of heparin the perfusate at the start of the EVLP circuit (approximately 1.5 IU/mL), and those after 6 hours of EVLP replenishment (approximately 0.65 IU/mL) are well below concentrations of heparin that would significantly inhibit PCR.


Example 6—Quantification of Human DNA in Ex Vivo Lung Perfusion (EVLP) Perfusates as Measured by a Reference Gene qPCR Method

Two mL volumes from initial Tubes 1-5 were subjected to automated DNA extraction after zero (Sample Level 1), one (Sample Level 2A), or two (Sample Level 3) 1100×g×10 min spins to remove cells and debris. The extracted DNA eluates were analyzed for total DNA concentration using a reference gene qPCR method. Results are shown in Table 4.









TABLE 4







Total Extracted DNA Concentration (ng/mL;


mean of duplicate determinations)









Processing Level (Refer to

Mean Total DNA (ng/ml


FIG. 3)
Tube Number
extracted)





1 (original unspun tube)
Tube 1
Undetectable



Tube 2
909



Tube 3
1924



Tube 4
856



Tube 5
1094


2a (supernatant after PPT
Tube 2
383


tube spin)
Tube 3
984



Tube 4
586



Tube 5
556


3 (supernatant after
Tube 2
462


additional 1100 g spin)
Tube 3
1052



Tube 4
429



Tube 5
472









No human DNA was detected in tube 1, consistent with no exposure within the perfusion circuit to a human lung. However, relatively high concentrations of human DNA (856-1924 ng/ml) were detected in tubes 2-5 prior to centrifugation (Level 1). With the progressive removal of intact nucleated cells from the fluid phase by centrifugation (Levels 2a and 3), the concentrations of total DNA present were predictably reduced, primarily as the result of the Level 2a spin that removed most of the cells, but remained relatively high in the cell-free supernatants of Levels 2a and 3.


The concentrations of cf-DNA observed in perfusate samples 2-5 (Levels 2a and 3) are notably elevated compared to, for instance, normal circulating cf-DNA levels historically observed in plasma from normal human subjects and most patients with heart transplant rejection. However, as noted in Example 7, the samples were shipped overnight without the essential spin within 2 hours of collection to separate cells from the fluid phase, where the true cf-DNA of interest is contained. For these current EVLP samples, for which centrifugation was not possible until after the overnight shipment was received, the high “cf-DNA” levels measured could have resulted from either post-collection leukocyte lysis or from cellular lysis occurring during EVLP.


Example 7—Cell Content of Perfusate Samples as a Function of Centrifugal Purification, Assessed by Automated Cell Count (Cell-Dyne)

Supernatant aliquots were collected from Tubes 1-5 after zero (level 1), one (level 2A), and two (level 3) 1100×g low speed spins. Cell count analyses on these aliquots were performed using a Cell-Dyne 3700 Hematology Analyzer (see Table 5).









TABLE 5







Cell count analyses (RBC/mL and WBC/mL) for Tubes 1-5












Processing Level
Tube





(Refer to FIG. 3)
Number
RBC/mL
WBC/mL







1 (original unspun
Tube 1
Undetectable
Undetectable



tube)
Tube 2
20670000
35640




Tube 3
27680000
43790




Tube 4
1800000
16840




Tube 5
1700000
16390



2a (supernatant
Tube 2
100000
4730



after PPT tube spin)
Tube 3
200000
2750




Tube 4
Undetectable
2640




Tube 5
Undetectable
3190



3 (supernatant after
Tube 2
Undetectable
Undetectable



additional 1100 g
Tube 3
100000
110



spin)
Tube 4
Undetectable
Undetectable




Tube 5
Undetectable
Undetectable










Cell count analysis shows close similarity, within the inherent variability of the Cell-Dyne method at these cell count levels, of Tubes 2 and 3, and of Tubes 4 and 5. These similarities are matched by comparisons of the appearances of the initially received tubes and the post-centrifugation tubes.


Importantly, centrifugation of the perfusate samples through levels 2a and 3 was found to be extremely effective in removing intact, countable cells. However, the slight residuum of RBCs and WBCs in one of the most cellular tubes (Tube 3) suggests a third spin may be beneficial prior to shipment for analysis.


Example 8—Evaluation of Cf-DNA Fragmentation in EVLP Perfusate Samples

DNA was extracted from duplicate 2 mL aliquots of pristine level 4 supernatants of Tubes 2-5 and a normal plasma control specimen using automated extraction technology. The extraction eluates were analyzed using a method for differentially detecting long and short fragments of DNA as a measure of the differential contributions of cellular apoptosis (the typical mode of cellular death in vivo that produces very short DNA fragments) versus cellular lysis typically occurring during sample processing (which produces longer DNA fragments). The proportion of long fragment DNA in the eluates (mean of duplicate determinations) is shown in FIG. 4.


Extracted DNA in the level 4, acellular supernatants prepared from UT tubes 2-5 contains a high proportion of long fragment DNA that exceeds the level which was previously determined to indicate a significant degree of leukocyte lysis which would contaminate the true cf-DNA complement of the fluid phase of the specimen with lysed leukocyte DNA. This would significantly complicate interpretation of cf-DNA levels as a measure of ex vivo lung injury during perfusion, unless those leukocytes and any other intact cells are removed quickly after sample collection by 2-3 short centrifugation steps at prior to shipment for analysis.


Example 9—Evaluation of Overall DNA Fragment Length Distribution in EVLP Perfusate Samples (Bioanalyzer Micro-Capillary-Based Electrophoresis)

Level 4 perfusate DNA from Tubes 2-5 was extracted by an automated extraction system. 1 μl of extracted eluate was loaded onto an Agilent High Sensitivity DNA Bioanalyzer chip, and run on TAI's Bioanalyzer 2100. This micro-capillary-based electrophoretic cell allows rapid and sensitive investigation of DNA fragment length distribution. A representative tracing is shown in FIG. 5.


The representative electropherogram and gel image in FIG. 5 shows a small peak of cf-DNA around 150 bp that is consistent with apoptotic DNA release, and a much larger peak extending from 500-15,000 bp. This 500-15,000 bp peak indicates marked cell lysis in the perfusate sample and independently confirms the findings of the long fragment proportion test (see Example 3).


Example 10—Evaluation of Recovery Efficiency of a Short DNA Fragment Control Spiked into Pre-EVLP STEEN™

25,000 copies of a short DNA fragment control, and 15 ng/ml sheared human gDNA (sheared to an average of 150 bp in length) were spiked into an aliquot of Tube 1 (level 1) fluid, which had been determined in Examples 1 and 2 to be acellular and without detectable endogenous DNA. DNA was extracted from this spiked sample of Tube 1 using an automated DNA extraction protocol, and the percent recovery of the short fragment DNA control and sheared gDNA in the DNA extraction eluate was determined using reference gene qPCR methods. Additionally, a short vs long DNA fragment assay was used to determine the DNA long fragment proportion of the spiked sample, which was expected to be low based on the shearing protocol employed in the experimental design. Results are shown below in Tables 6-8.









TABLE 6







Percent Recovery of Short Fragment DNA Control after Extraction









Processing Level (Refer to
Sample
Mean Short Fragment DNA


FIG. 3)
Name
Control Recovery %





1
Tube 1
47%
















TABLE 7







Percent Recovery of Spiked Sheared gDNA after Extraction











Processing Level (Refer to

Mean Sheared gDNA



FIG. 3)
Sample Name
Recovery %







1
Tube 1
50%

















TABLE 8







Long Fragment DNA Proportion of Spiked


Sheared gDNA after Extraction











Processing Level

Mean Sheared gDNA Long



(Refer to FIG. 3)
Sample Name
Fragment Proportion







1
Tube 1
0.10










The data presented in Tables 6-8 demonstrates that the automated cf-DNA extraction methodology utilized performs well in extracting short DNA fragments (which are typical of apoptotic cf-DNA) from pre-EVLP STEEN solution-containing standard EVLP additives. The extraction efficiency is comparable to that historically seen when extracting short fragment DNA from human plasma samples using the same method. As a general rule, short DNA fragments are not as easily extracted as long DNA fragments by any DNA extraction method, but the instant procedure has been optimized to do this well and it does so from STEEN solutions as well as from plasma.


Example 11—Immunophenotypic Identification of Contaminating Cell Types in EVLP Perfusate Samples (Flow Cytometry)

Five ml aliquots from level 1 Tubes 2-5 were transferred per the sample processing protocol shown in FIG. 3 into 15 ml conical tubes and centrifuged for 10 minutes at 1100×g (Level 2b). Supernatants were removed and pellets were resuspended in 500 μl PBS for flow cytometric analysis. Results are shown below for each tube.












Tube 2.










Population
#Events
% Parent
% Total













custom character   All Events
13,698
####
100.0


custom character   WBCs
6,402
46.7
46.7


custom character   Lymphocytes
2,099
32.8
15.3


custom character   CD4+ CD3+
323
15.4
2.4


custom character   CD8+ CD3+
369
17.6
2.7


custom character   CD56+
1,144
54.5
8.4


custom character   CD56+ CD3+
44
2.1
0.3


custom character   CD3+
677
32.3
4.9


custom character   CD19+
155
7.4
1.1


custom character   Monocytes
172
2.7
1.3


custom character   Granulocytes
2,288
35.7
16.7



















Tube 3.












Population
#Events
% Parent
% Total
















custom character   All Events
22,560
####
100.0



custom character   WBCs
12,419
55.0
55.0



custom character   Lymphocytes
4,127
33.2
18.3



custom character   CD4+ CD3+
884
21.4
3.9



custom character   CD8+ CD3+
654
15.8
2.9



custom character   CD56+
1,790
43.4
7.9



custom character   CD56+ CD3+
65
1.6
0.3



custom character   CD3+
1,526
37.0
6.8



custom character   CD19+
591
14.3
2.6



custom character   Monocytes
589
4.7
2.6



custom character   Granulocytes
4,693
37.8
20.8




















Tube 4.










Population
#Events
% Parent
% Total













custom character   All Events
14,601
####
100.0


custom character   WBCs
6,928
47.4
47.4


custom character   Lymphocytes
2,323
33.5
15.9


custom character   CD4+ CD3+
910
39.2
6.2


custom character   CD8+ CD3+
434
18.7
3.0


custom character   CD56+
546
23.5
3.7


custom character   CD56+ CD3+
55
2.4
0.4


custom character   CD3+
1,316
56.7
9.0


custom character   CD19+
283
12.2
1.9


custom character   Monocytes
112
1.6
0.8


custom character   Granulocytes
2,547
36.8
17.4



















Tube 5.










Population
#Events
% Parent
% Total













custom character   All Events
13,806
####
100.0


custom character   WBCs
5,970
43.2
43.2


custom character   Lymphocytes
2,272
38.1
16.5


custom character   CD4+ CD3+
881
38.8
6.4


custom character   CD8+ CD3+
442
19.5
3.2


custom character   CD56+
564
24.8
4.1


custom character   CD56+ CD3+
57
2.5
0.4


custom character   CD3+
1,304
57.4
9.4


custom character   CD19+
264
11.6
1.9


custom character   Monocytes
105
1.8
0.8


custom character   Granulocytes
2,072
34.7
15.0










The tables generated by the flow cytometry analysis for Tubes 2-5 show the samples to be comparable in cellular composition as revealed by a standard human leukocyte antibody panel. This is consistent with the cellular morphology seen in matching cytospin preparations.


Example 12—Materials and Methods (Examples 6-11)

Five de-identified, uncentrifuged samples from human ex vivo lung perfusion (EVLP) procedures were obtained (initial Tubes 1-5). The EVLP procedure used included a gradual rewarming of the lungs to normal core body temperature in conjunction with a gradual increase in vascular flow, targeting a perfusion flow of 40% donor-predicted cardiac output (CO) (Machuca et al., J Thorac Dis. 2014, 6(8):1054-1106). Protective lung ventilation and an acellular perfusate with increased colloid osmotic pressure were attained through the use of human serum albumin and Dexran 40. The methodology has been FDA-approved under a humanitarian device exemption (HDE). During EVLP, the perfusion circuit of the lung mimics in vivo conditions. The ventilated ex vivo lungs are perfused with STEEN™ solution without red blood cells. Parameters, such as gaseous exchange, pulmonary vascular resistance, compliance, and other key variables under normothermic conditions are monitored. Six hours or more of EVLP is clinically considered the standard when using an acellular STEEN perfusate. STEEN™ solution, a buffered extracellular solution includes human serum albumin for osmotic pressure and Dextran 40, a mild scavenger used to coat and protect the endothelium from excessive leukocyte interaction (Steen et al., Lancet 2001, 357:825-829; Steen et al., Ann Thorac Surg. 2003, 76:244-252; Steen et al., Ann Thorac Surg. 2007; 83:2191). The solution is designed to facilitate prolonged evaluation of lung transplantation options and to promote health of the isolated lungs ex vivo. EVLP using the STEEN™ solution thus has the potential to be able to increase the likelihood that previously rejected, but ex vivo rehabilitated lungs could be used to increase the availability of potential organs for lung transplantation.


The five samples were collected from two human lung perfusion procedures. The samples were shipped, unprocessed, overnight with cold packs. The samples were never frozen. Sample details are provided in Table 9 below.









TABLE 9







EVLP Sample Collection Details












Tube#
1
2
3
4
5





Amount
 30 ml
 30 ml
 30 ml
 30 ml
 30 ml


Perfusate type
LPD-2A (Steen)
Single right
Single right
Single right
Single right


Timing
Before Lungs
After 1 hour
After 2 hour perfusion,
After 1 hour
After 2 hour



perfusion. LPD-2A
perfusion,
before exchange
perfusion, before
perfusion, before



was Primed in
before

exchange
exchange



circuit. No contact
exchange






to lungs.






Prime amount
2000 ml
2000 ml
2000 ml
2000 ml
2000 ml


Additional
Heparin 3000 IU,

500 ml exchange after

500 ml exchange


information
Imipenum 500 mg,

sampling

after sampling



Solumedrol 500 mg









The five samples were subjected to the centrifugation steps outlined in FIG. 3. The resulting supernatants and cellular pellets were used in the Experiments described below.


For the cellular analyses, as indicated in the right half of FIG. 3 (level 2B), 5 mL aliquots from each of four received tubes (Tubes 2-5 from level 1) were transferred into two individual 15 mL conical tubes spiked with either a cellular preservative or no preservative. Tubes were centrifuged at 1100×g for 10 minutes. Tubes 2 and 3 and Tubes 4 and 5 showed similar appearances. The resulting level 2b supernatants were removed, and the remaining cell pellets were resuspended in 0.5 mL phosphate buffered saline (PBS) for flow cytometry and cell counting.


For cf-DNA extraction and fragment analysis, shown as level 2a in FIG. 3, 8.5 mL aliquots from each of the received tubes (Tubes 1-5 of level 1) were transferred from those tubes into similarly labeled individual PPT tubes. The PPT tubes were then centrifuged at 1100×g for 10 minutes. Tube 1 yielded no cellular pellet. A 500 μL aliquot of each level 2a supernatant was collected for cell counting analysis (Cell Dyne) and the remaining approximately 8 mL of supernatant was transferred to a fresh 15 mL conical tube that was then subjected to a second low speed spin at 1100×g for 10 minutes (level 3 of FIG. 3). There was a lack of a visually obvious residual cellular pellet following this spin step.


Aliquots of the level 3 supernatants were collected (500 μL) for cell count analysis, flow cytometer, and initial RNAseP DNA quantification. Avoiding the last 500 μL in the tube's bottom tip, the remaining supernatants were transferred to a fresh 15 mL conical tubes and centrifuged at 15,000×g for 15 minutes. The resultant pristine level 4 supernatants (FIG. 3) were collected for cf-DNA extraction and quantification/fragment analysis.

Claims
  • 1. A method of preparing a preparation of amplified DNA from a sample comprising DNA released from a potential graft and assessing the suitability of the potential graft for transplantation or implantation, comprising: (a) extracting cell-free DNA (cf-DNA) from the sample, wherein the sample is obtained from a perfusate in contact with the potential graft ex vivo and comprises cf-DNA released from the potential graft subsequent to contacting of the potential graft with blood cells from a potential recipient, wherein the perfusate is a buffered extracellular solution comprising Dextran 40 and human serum albumin, wherein the potential graft is a lung, and wherein the sample is processed with a step that removes contaminating cells from the sample prior to extracting cf-DNA,(b) amplifying the cf-DNA extracted in (a), and(c) assessing the amplified DNA to quantify amounts of both total cf-DNA and graft-specific cf-DNA to determine the suitability of the potential graft for transplantation or implantation, andwherein the method further comprises quantifying short DNA fragments having 75-170 base pairs in length which are indicative of cell apoptosis and long DNA fragments having 500-15,000 base pairs in length which are indicative of cell lysis.
  • 2. The method of claim 1, wherein the method further comprises obtaining the potential graft.
  • 3. The method of claim 1, wherein the method further comprises obtaining blood from a potential recipient.
  • 4. The method of claim 1, wherein the method further comprises obtaining amounts of total cf-DNA and graft-specific cf-DNA at one or more additional time points.
  • 5. The method of claim 1, wherein the method further comprises comparing the amounts of the total cf-DNA and graft-specific cf-DNA with threshold values of total cf-DNA and graft-specific cf-DNA obtained from one or more additional time points.
  • 6. The method of claim 1, wherein the potential graft is monitored over time.
  • 7. The method of claim 1, wherein step (c) comprises quantifying the amounts of total cf-DNA and graft-specific cf-DNA by performing real-time PCR or digital PCR.
  • 8. The method of claim 1, wherein step (c) comprises quantifying the amounts of the total cf-DNA and graft-specific cf-DNA by performing next-generation sequencing.
  • 9. The method of claim 1, wherein the amounts of total cf-DNA and graft-specific cf-DNA above a threshold value of total cell-free DNA and graft-specific cf-DNA determined from a different point in time is indicative of decreasing suitability, or unsuitability, of the graft.
  • 10. The method of claim 1, wherein the amounts of total cf-DNA and graft-specific cf-DNA below a threshold value of total cell-free DNA and graft-specific cf-DNA determined from a different point in time is indicative of increasing suitability, or suitability, of the graft.
  • 11. The method of claim 1, wherein step (b) comprises performing mismatch PCR amplification to obtain the preparation of amplified DNA.
  • 12. The method of claim 1, wherein the potential graft is obtained from a different species than human.
  • 13. The method of claim 1, wherein the potential graft is obtained from a pig.
RELATED APPLICATION

This application is a national stage filing under 35 U.S.C. § 371 of International Patent Application No. PCT/US2018/065845, filed Dec. 14, 2018, which claims the benefit under 35 U.S.C. § 119 of the filing date of U.S. Provisional Application No. 62/599,011, filed Dec. 14, 2017, the contents of each of which are incorporated herein by reference in their entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2018/065845 12/14/2018 WO
Publishing Document Publishing Date Country Kind
WO2019/118926 6/20/2019 WO A
US Referenced Citations (694)
Number Name Date Kind
3957654 Ayres May 1976 A
4040785 Kim et al. Aug 1977 A
4683195 Mullis et al. Jul 1987 A
4942124 Church et al. Jul 1990 A
5180812 Dower et al. Jan 1993 A
5314809 Erlich et al. May 1994 A
5319071 Dower et al. Jun 1994 A
5464937 Sims et al. Nov 1995 A
5486477 Carver Jan 1996 A
5488032 Dower et al. Jan 1996 A
5492888 Dower et al. Feb 1996 A
5569582 Tavernarakis et al. Oct 1996 A
5595890 Newton et al. Jan 1997 A
5635366 Cooke et al. Jun 1997 A
5645988 Vande Woude et al. Jul 1997 A
5648220 Bianchi Jul 1997 A
5714320 Kool Feb 1998 A
5716776 Bogart Feb 1998 A
5736033 Coleman et al. Apr 1998 A
5753467 Jensen et al. May 1998 A
5824467 Mascarenhas Oct 1998 A
5854033 Lizardi Dec 1998 A
5860917 Comanor et al. Jan 1999 A
5891734 Gill et al. Apr 1999 A
5952170 Stroun et al. Sep 1999 A
5962223 Whiteley et al. Oct 1999 A
5972602 Hyland et al. Nov 1999 A
5976790 Pinkel et al. Nov 1999 A
5994148 Stewart et al. Nov 1999 A
6001611 Will Dec 1999 A
6025128 Veltri et al. Feb 2000 A
6066454 Lipshutz et al. May 2000 A
6100029 Lapidus et al. Aug 2000 A
6108635 Herren et al. Aug 2000 A
6124120 Lizardi Sep 2000 A
6143496 Brown et al. Nov 2000 A
6156504 Gocke et al. Dec 2000 A
6180349 Ginzinger Jan 2001 B1
6214558 Shuber et al. Apr 2001 B1
6221603 Mahtani Apr 2001 B1
6235472 Landegren et al. May 2001 B1
6258540 Lo et al. Jul 2001 B1
6300077 Shuber et al. Oct 2001 B1
6329179 Kopreski Dec 2001 B1
6335167 Pinkel et al. Jan 2002 B1
6440706 Vogelstein et al. Aug 2002 B1
6479235 Schumm et al. Nov 2002 B1
6489135 Parrott et al. Dec 2002 B1
6605451 Marmaro et al. Aug 2003 B1
6617137 Dean et al. Sep 2003 B2
6720140 Hartley et al. Apr 2004 B1
6794140 Goldsborough Sep 2004 B1
6807491 Pavlovic et al. Oct 2004 B2
6852487 Barany et al. Feb 2005 B1
6858412 Willis et al. Feb 2005 B2
6927028 Lo et al. Aug 2005 B2
6958211 Vingerhoets et al. Oct 2005 B2
6964847 Englert Nov 2005 B1
7035739 Schadt et al. Apr 2006 B2
7058517 Denton et al. Jun 2006 B1
7058616 Larder et al. Jun 2006 B1
7101663 Godfrey et al. Sep 2006 B2
7153656 Nolan et al. Dec 2006 B2
7218764 Vaisberg et al. May 2007 B2
7297485 Bornarth et al. Nov 2007 B2
7332277 Dhallan Feb 2008 B2
7410764 Gocke et al. Aug 2008 B2
7414118 Mullah et al. Aug 2008 B1
7442506 Dhallan Oct 2008 B2
7459273 Jones et al. Dec 2008 B2
7645576 Lo et al. Jan 2010 B2
7655399 Cantor et al. Feb 2010 B2
7700325 Cantor et al. Apr 2010 B2
7718367 Lo et al. May 2010 B2
7718370 Dhallan May 2010 B2
7727720 Dhallan Jun 2010 B2
7741463 Gormley et al. Jun 2010 B2
7785798 Cantor et al. Aug 2010 B2
7790393 Lyamichev et al. Sep 2010 B2
7790418 Mayer Sep 2010 B2
7805282 Casey Sep 2010 B2
7838647 Hahn et al. Nov 2010 B2
7888017 Quake Feb 2011 B2
7981609 Rubin et al. Jul 2011 B2
8008018 Quake et al. Aug 2011 B2
8024128 Rabinowitz et al. Sep 2011 B2
8133719 Drmanac et al. Mar 2012 B2
8137912 Kapur et al. Mar 2012 B2
8168389 Shoemaker et al. May 2012 B2
8173370 Oeth et al. May 2012 B2
8195415 Fan et al. Jun 2012 B2
8236503 Faham et al. Aug 2012 B2
8296076 Fan et al. Oct 2012 B2
8304187 Fernando Nov 2012 B2
8318430 Chuu et al. Nov 2012 B2
8318434 Cuppens et al. Nov 2012 B2
8323897 Andersen et al. Dec 2012 B2
8372584 Shoemaker et al. Feb 2013 B2
8389557 Singh et al. Mar 2013 B2
8389578 Went et al. Mar 2013 B2
8450063 Dube et al. May 2013 B2
8467976 Lo et al. Jun 2013 B2
8515679 Rabinowitz et al. Aug 2013 B2
8532930 Rabinowitz et al. Sep 2013 B2
8609338 Mitchell et al. Dec 2013 B2
8679741 Hoyal-Wrightson et al. Mar 2014 B2
8682592 Rabinowitz et al. Mar 2014 B2
8703652 Quake et al. Apr 2014 B2
8706422 Lo et al. Apr 2014 B2
8748103 Faham et al. Jun 2014 B2
8822153 Hayes et al. Sep 2014 B2
8825412 Rabinowitz et al. Sep 2014 B2
9005894 Ladner et al. Apr 2015 B2
9051602 Oliphant et al. Jun 2015 B2
9085798 Chee Jul 2015 B2
9206475 Gerdes et al. Dec 2015 B2
9228234 Rabinowitz et al. Jan 2016 B2
9290815 Di Pasquale et al. Mar 2016 B2
9323888 Rava et al. Apr 2016 B2
9364829 Heid et al. Jun 2016 B2
9404150 Lee et al. Aug 2016 B2
9424392 Rabinowitz et al. Aug 2016 B2
9453257 Hoyal-Wrightson et al. Sep 2016 B2
9476095 Vogelstein et al. Oct 2016 B2
9487829 Vogelstein et al. Nov 2016 B2
9493828 Rava et al. Nov 2016 B2
9506119 Faham et al. Nov 2016 B2
9598731 Talasaz Mar 2017 B2
9677118 Zimmermann et al. Jun 2017 B2
9926593 Ehrich et al. Mar 2018 B2
9957558 Leamon et al. May 2018 B2
10011870 Zimmermann et al. Jul 2018 B2
10017810 Iafrate et al. Jul 2018 B2
10041127 Talasaz Aug 2018 B2
10061890 Rabinowitz et al. Aug 2018 B2
10081839 Rabinowitz et al. Sep 2018 B2
10083273 Rabinowitz et al. Sep 2018 B2
10174369 Rabinowitz et al. Jan 2019 B2
10179937 Babiarz et al. Jan 2019 B2
10227652 Rabinowitz et al. Mar 2019 B2
10229244 Ghosh Mar 2019 B2
10240202 Rabinowitz et al. Mar 2019 B2
10260096 Rabinowitz et al. Apr 2019 B2
10266893 Rabinowitz et al. Apr 2019 B2
10308981 Sparks et al. Jun 2019 B2
10316362 Babiarz et al. Jun 2019 B2
10351906 Zimmermann et al. Jul 2019 B2
10385396 Mitchell et al. Aug 2019 B2
10392664 Rabinowitz et al. Aug 2019 B2
10450597 Iafrate et al. Oct 2019 B2
10472680 Mitchell et al. Nov 2019 B2
10522242 Rabinowitz et al. Dec 2019 B2
10526658 Babiarz et al. Jan 2020 B2
10538814 Babiarz et al. Jan 2020 B2
10557172 Babiarz et al. Feb 2020 B2
10597708 Zimmermann et al. Mar 2020 B2
10597709 Zimmermann et al. Mar 2020 B2
10597723 Babiarz et al. Mar 2020 B2
10655180 Babiarz et al. May 2020 B2
10683552 Giulio et al. Jun 2020 B2
10711309 Rabinowitz et al. Jul 2020 B2
10731220 Babiarz et al. Aug 2020 B2
10774380 Ryan et al. Sep 2020 B2
10793912 Babiarz et al. Oct 2020 B2
10894976 Stray et al. Jan 2021 B2
11111543 Rabinowitz et al. Sep 2021 B2
11111544 Rabinowitz et al. Sep 2021 B2
11111545 Babiarz et al. Sep 2021 B2
11130995 Quake et al. Sep 2021 B2
20010051341 Lo et al. Dec 2001 A1
20010053519 Fodor et al. Dec 2001 A1
20020006622 Bradley et al. Jan 2002 A1
20020107640 Ideker et al. Aug 2002 A1
20020119478 Umansky et al. Aug 2002 A1
20020182622 Nakamura et al. Dec 2002 A1
20030009295 Markowitz et al. Jan 2003 A1
20030040620 Langmore et al. Feb 2003 A1
20030044388 Lo et al. Mar 2003 A1
20030065535 Karlov et al. Apr 2003 A1
20030077586 Pavlovic et al. Apr 2003 A1
20030087276 Kopreski et al. May 2003 A1
20030101000 Bader et al. May 2003 A1
20030108900 Oliphant et al. Jun 2003 A1
20030119004 Wenz et al. Jun 2003 A1
20030138780 Gill et al. Jul 2003 A1
20030148301 Aono et al. Aug 2003 A1
20030191005 Coelho et al. Oct 2003 A1
20030211489 Shen et al. Nov 2003 A1
20030228613 Bornarth et al. Dec 2003 A1
20030232348 Jones et al. Dec 2003 A1
20030232353 Kennedy et al. Dec 2003 A1
20030235848 Neville et al. Dec 2003 A1
20040009518 Lo et al. Jan 2004 A1
20040033596 Threadgill et al. Feb 2004 A1
20040067493 Matsuzaki et al. Apr 2004 A1
20040096874 Neville et al. May 2004 A1
20040115629 Panzer et al. Jun 2004 A1
20040117346 Stoffel et al. Jun 2004 A1
20040126760 Broude Jul 2004 A1
20040136967 Weiss et al. Jul 2004 A1
20040137470 Dhallan et al. Jul 2004 A1
20040146866 Fu Jul 2004 A1
20040157243 Huang et al. Aug 2004 A1
20040185495 Schueler et al. Sep 2004 A1
20040197797 Inoko et al. Oct 2004 A1
20040209299 Pinter et al. Oct 2004 A1
20040229231 Frudakis et al. Nov 2004 A1
20040236518 Pavlovic et al. Nov 2004 A1
20040259100 Gunderson et al. Dec 2004 A1
20050009069 Liu et al. Jan 2005 A1
20050043894 Fernandez Feb 2005 A1
20050049793 Paterlini-brechot Mar 2005 A1
20050053950 Ubani et al. Mar 2005 A1
20050064476 Huang et al. Mar 2005 A1
20050079521 Beaulieu et al. Apr 2005 A1
20050079535 Kirchgesser et al. Apr 2005 A1
20050123914 Katz et al. Jun 2005 A1
20050130173 Leamon et al. Jun 2005 A1
20050142577 Jones et al. Jun 2005 A1
20050144664 Smith et al. Jun 2005 A1
20050164241 Hahn et al. Jul 2005 A1
20050164252 Yeung Jul 2005 A1
20050216207 Kermani Sep 2005 A1
20050221341 Shimkets et al. Oct 2005 A1
20050227263 Green et al. Oct 2005 A1
20050250111 Xie et al. Nov 2005 A1
20050255508 Casey et al. Nov 2005 A1
20050272073 Vaisberg et al. Dec 2005 A1
20050282185 Lo et al. Dec 2005 A1
20060014179 Roberts Jan 2006 A1
20060019278 Lo et al. Jan 2006 A1
20060040300 Dapprich et al. Feb 2006 A1
20060046258 Lapidus et al. Mar 2006 A1
20060051799 Iwaki et al. Mar 2006 A1
20060052945 Rabinowitz et al. Mar 2006 A1
20060057618 Piper et al. Mar 2006 A1
20060068369 Coelho et al. Mar 2006 A1
20060068394 Langmore et al. Mar 2006 A1
20060088574 Manning et al. Apr 2006 A1
20060088871 Finkelstein et al. Apr 2006 A1
20060088912 Yan et al. Apr 2006 A1
20060094010 Giles et al. May 2006 A1
20060099614 Gill et al. May 2006 A1
20060121452 Dhallan Jun 2006 A1
20060134662 Pratt et al. Jun 2006 A1
20060141499 Sher et al. Jun 2006 A1
20060210997 Myerson et al. Sep 2006 A1
20060216153 Wobben et al. Sep 2006 A1
20060216738 Wada et al. Sep 2006 A1
20060228721 Leamon et al. Oct 2006 A1
20060229823 Liu Oct 2006 A1
20060234264 Hardenbol Oct 2006 A1
20060248031 Kates et al. Nov 2006 A1
20060281105 Li et al. Dec 2006 A1
20060292599 Ritz et al. Dec 2006 A1
20070020640 McCloskey et al. Jan 2007 A1
20070027636 Rabinowitz Feb 2007 A1
20070031857 Makarov et al. Feb 2007 A1
20070037166 Wohlgemuth et al. Feb 2007 A1
20070042384 Li et al. Feb 2007 A1
20070059700 Tao et al. Mar 2007 A1
20070059707 Cantor et al. Mar 2007 A1
20070122805 Cantor et al. May 2007 A1
20070128624 Gormley et al. Jun 2007 A1
20070134658 Bohmer et al. Jun 2007 A1
20070178478 Dhallan Aug 2007 A1
20070178501 Rabinowitz et al. Aug 2007 A1
20070184467 Rabinowitz et al. Aug 2007 A1
20070202525 Quake et al. Aug 2007 A1
20070202536 Yamanishi et al. Aug 2007 A1
20070207466 Cantor et al. Sep 2007 A1
20070212689 Bianchi et al. Sep 2007 A1
20070231823 Mckernan et al. Oct 2007 A1
20070243549 Bischoff Oct 2007 A1
20070259351 Chinitz Nov 2007 A1
20080020390 Mitchell Jan 2008 A1
20080026390 Stoughton et al. Jan 2008 A1
20080038733 Bischoff et al. Feb 2008 A1
20080050739 Stoughton et al. Feb 2008 A1
20080070792 Stoughton Mar 2008 A1
20080071076 Hahn et al. Mar 2008 A1
20080085836 Kearns et al. Apr 2008 A1
20080090239 Shoemaker et al. Apr 2008 A1
20080096766 Lee Apr 2008 A1
20080102455 Poetter May 2008 A1
20080138809 Kapur et al. Jun 2008 A1
20080161420 Shuber et al. Jul 2008 A1
20080164204 Hatamian et al. Jul 2008 A1
20080182244 Tafas et al. Jul 2008 A1
20080193927 Mann et al. Aug 2008 A1
20080220422 Shoemaker et al. Sep 2008 A1
20080234142 Lietz Sep 2008 A1
20080243398 Rabinowitz et al. Oct 2008 A1
20080280292 Wangh et al. Nov 2008 A1
20080286783 Hosono et al. Nov 2008 A1
20080299562 Oeth et al. Dec 2008 A1
20080305473 Chowdary et al. Dec 2008 A1
20090023190 Lao et al. Jan 2009 A1
20090029377 Lo et al. Jan 2009 A1
20090053719 Lo et al. Feb 2009 A1
20090087847 Lo et al. Apr 2009 A1
20090098534 Weier et al. Apr 2009 A1
20090099041 Church et al. Apr 2009 A1
20090143570 Jiang et al. Jun 2009 A1
20090176234 Drmanac et al. Jul 2009 A1
20090176662 Rigatti et al. Jul 2009 A1
20090221620 Luke et al. Sep 2009 A1
20090228299 Kangarloo et al. Sep 2009 A1
20090233802 Bignell et al. Sep 2009 A1
20090253183 Han Oct 2009 A1
20090263800 Wohlgemuth et al. Oct 2009 A1
20090280479 Hoon et al. Nov 2009 A1
20090317817 Oeth et al. Dec 2009 A1
20100012598 Dicesare et al. Jan 2010 A1
20100035232 Ecker et al. Feb 2010 A1
20100041048 Diehl et al. Feb 2010 A1
20100086914 Bentley et al. Apr 2010 A1
20100105049 Ehrich et al. Apr 2010 A1
20100112575 Fan et al. May 2010 A1
20100112586 Stoughton et al. May 2010 A1
20100112590 Lo et al. May 2010 A1
20100120038 Mir et al. May 2010 A1
20100124751 Quake et al. May 2010 A1
20100129792 Makrigiorgos et al. May 2010 A1
20100129874 Mitra et al. May 2010 A1
20100138165 Fan et al. Jun 2010 A1
20100155343 Battles et al. Jun 2010 A1
20100171954 Quake et al. Jul 2010 A1
20100173394 Colston et al. Jul 2010 A1
20100184043 Mitchell et al. Jul 2010 A1
20100184069 Fernando et al. Jul 2010 A1
20100184152 Sandler Jul 2010 A1
20100196892 Quake et al. Aug 2010 A1
20100203538 Dube et al. Aug 2010 A1
20100216145 Duvdevani Aug 2010 A1
20100216151 Lapidus et al. Aug 2010 A1
20100216153 Lapidus et al. Aug 2010 A1
20100227329 Cuppens Sep 2010 A1
20100248231 Wei et al. Sep 2010 A1
20100255492 Quake et al. Oct 2010 A1
20100256013 Quake et al. Oct 2010 A1
20100273159 Melo Oct 2010 A1
20100273219 May et al. Oct 2010 A1
20100273678 Alexandre et al. Oct 2010 A1
20100285478 Chen et al. Nov 2010 A1
20100285537 Zimmermann Nov 2010 A1
20100291572 Stoughton et al. Nov 2010 A1
20100291635 Peleg Nov 2010 A1
20100323352 Lo et al. Dec 2010 A1
20100326218 Boeckh et al. Dec 2010 A1
20110015096 Chiu Jan 2011 A1
20110033862 Rabinowitz et al. Feb 2011 A1
20110039724 Lo et al. Feb 2011 A1
20110045462 Fu et al. Feb 2011 A1
20110064824 Lascoste et al. Mar 2011 A1
20110071031 Khripin et al. Mar 2011 A1
20110086769 Oliphant et al. Apr 2011 A1
20110092763 Rabinowitz et al. Apr 2011 A1
20110105353 Lo et al. May 2011 A1
20110110931 Matsui May 2011 A1
20110111410 Ryan et al. May 2011 A1
20110130558 Ritt et al. Jun 2011 A1
20110151442 Fan et al. Jun 2011 A1
20110159499 Hindson et al. Jun 2011 A1
20110160078 Fodor et al. Jun 2011 A1
20110178719 Rabinowitz et al. Jul 2011 A1
20110189677 Adli et al. Aug 2011 A1
20110201507 Rava et al. Aug 2011 A1
20110212446 Wang et al. Sep 2011 A1
20110212846 Spier Sep 2011 A1
20110224087 Quake et al. Sep 2011 A1
20110230358 Rava Sep 2011 A1
20110246083 Fan et al. Oct 2011 A1
20110251149 Perrine et al. Oct 2011 A1
20110288780 Rabinowitz et al. Nov 2011 A1
20110294699 Lee et al. Dec 2011 A1
20110300608 Ryan et al. Dec 2011 A1
20110301854 Curry et al. Dec 2011 A1
20110312503 Chuu et al. Dec 2011 A1
20110318734 Lo et al. Dec 2011 A1
20120003635 Lo et al. Jan 2012 A1
20120003637 Lo et al. Jan 2012 A1
20120010085 Rava et al. Jan 2012 A1
20120021442 Buhimschi et al. Jan 2012 A1
20120028814 Toloue et al. Feb 2012 A1
20120034603 Oliphant et al. Feb 2012 A1
20120034685 Sparks et al. Feb 2012 A1
20120108460 Quake et al. May 2012 A1
20120115140 Rivkees et al. May 2012 A1
20120122701 Ryan et al. May 2012 A1
20120122702 Leproust et al. May 2012 A1
20120135872 Chuu et al. May 2012 A1
20120165203 Quake et al. Jun 2012 A1
20120185176 Rabinowitz et al. Jul 2012 A1
20120190020 Oliphant et al. Jul 2012 A1
20120190021 Oliphant et al. Jul 2012 A1
20120190557 Oliphant et al. Jul 2012 A1
20120191358 Oliphant et al. Jul 2012 A1
20120196754 Quake et al. Aug 2012 A1
20120208706 Downing et al. Aug 2012 A1
20120214678 Rava et al. Aug 2012 A1
20120251411 Jeon Oct 2012 A1
20120264121 Rava et al. Oct 2012 A1
20120264618 Nygren Oct 2012 A1
20120270212 Rabinowitz et al. Oct 2012 A1
20120270739 Rava et al. Oct 2012 A1
20120295810 Quake et al. Nov 2012 A1
20120295819 Leamon et al. Nov 2012 A1
20130017549 Hong Jan 2013 A1
20130022973 Hansen et al. Jan 2013 A1
20130024127 Stuelpnagel Jan 2013 A1
20130034546 Rava et al. Feb 2013 A1
20130040375 Sparks et al. Feb 2013 A1
20130060483 Struble et al. Mar 2013 A1
20130069869 Akao et al. Mar 2013 A1
20130071844 Makino et al. Mar 2013 A1
20130085681 Deciu et al. Apr 2013 A1
20130090250 Sparks et al. Apr 2013 A1
20130116130 Fu et al. May 2013 A1
20130123120 Zimmermann et al. May 2013 A1
20130130923 Ehrich et al. May 2013 A1
20130143219 Mitchell et al. Jun 2013 A1
20130157870 Pushkarev et al. Jun 2013 A1
20130172211 Oliphant et al. Jul 2013 A1
20130178373 Rabinowitz et al. Jul 2013 A1
20130190653 Alvarez Ramos Jul 2013 A1
20130196862 Rabinowitz et al. Aug 2013 A1
20130210644 Stoughton et al. Aug 2013 A1
20130225422 Rabinowitz et al. Aug 2013 A1
20130231252 Mitchell et al. Sep 2013 A1
20130237431 Lo et al. Sep 2013 A1
20130252824 Rabinowitz Sep 2013 A1
20130253369 Rabinowitz et al. Sep 2013 A1
20130261004 Ryan et al. Oct 2013 A1
20130274116 Rabinowitz et al. Oct 2013 A1
20130274135 Zhang et al. Oct 2013 A1
20130288252 Sparks et al. Oct 2013 A1
20130303461 Iafrate et al. Nov 2013 A1
20130310260 Kim et al. Nov 2013 A1
20130323727 Huang et al. Dec 2013 A1
20130323731 Lo et al. Dec 2013 A1
20130325360 Deciu et al. Dec 2013 A1
20130344066 Faham et al. Dec 2013 A1
20140032128 Rabinowitz et al. Jan 2014 A1
20140038830 Srinivasan et al. Feb 2014 A1
20140045181 Lo et al. Feb 2014 A1
20140051585 Prosen et al. Feb 2014 A1
20140065621 Mhatre et al. Mar 2014 A1
20140066317 Talasaz Mar 2014 A1
20140087385 Rabinowitz et al. Mar 2014 A1
20140094373 Zimmermann et al. Apr 2014 A1
20140100121 Lo et al. Apr 2014 A1
20140100126 Rabinowitz Apr 2014 A1
20140100134 Rabinowitz et al. Apr 2014 A1
20140106975 Stoughton et al. Apr 2014 A1
20140113795 Emerson et al. Apr 2014 A1
20140141981 Zimmermann et al. May 2014 A1
20140154682 Rabinowitz et al. Jun 2014 A1
20140155274 Xie et al. Jun 2014 A1
20140162269 Rabinowitz Jun 2014 A1
20140186827 Pieprzyk et al. Jul 2014 A1
20140193816 Rabinowitz et al. Jul 2014 A1
20140206552 Rabinowitz et al. Jul 2014 A1
20140227691 May et al. Aug 2014 A1
20140227705 Vogelstein et al. Aug 2014 A1
20140242588 Van Den Boom et al. Aug 2014 A1
20140256558 Varley et al. Sep 2014 A1
20140256569 Rabinowitz et al. Sep 2014 A1
20140272956 Huang et al. Sep 2014 A1
20140274740 Srinivasan et al. Sep 2014 A1
20140287934 Szelinger et al. Sep 2014 A1
20140296081 Diehn et al. Oct 2014 A1
20140329245 Spier et al. Nov 2014 A1
20140336060 Rabinowitz Nov 2014 A1
20150051087 Rabinowitz et al. Feb 2015 A1
20150056617 Whitt et al. Feb 2015 A1
20150064695 Katz et al. Mar 2015 A1
20150086477 Mitchell et al. Mar 2015 A1
20150087535 Patel Mar 2015 A1
20150099673 Fodor Apr 2015 A1
20150147815 Babiarz et al. May 2015 A1
20150167069 Schutz et al. Jun 2015 A1
20150167077 Fehr et al. Jun 2015 A1
20150197786 Osborne et al. Jul 2015 A1
20150211050 Iafrate et al. Jul 2015 A1
20150218631 Chuu et al. Aug 2015 A1
20150232938 Mhatre Aug 2015 A1
20150246103 Hazout Sep 2015 A1
20150265995 Head et al. Sep 2015 A1
20150299812 Talasaz Oct 2015 A1
20150315657 Rhodes et al. Nov 2015 A1
20150322507 Zimmermann et al. Nov 2015 A1
20150329891 Tan et al. Nov 2015 A1
20160024581 Sarwal et al. Jan 2016 A1
20160032396 Diehn et al. Feb 2016 A1
20160046986 Eltoukhy et al. Feb 2016 A1
20160053320 Schuh et al. Feb 2016 A1
20160115541 Schutz et al. Apr 2016 A1
20160145682 Woodward et al. May 2016 A1
20160186239 Sinha Jun 2016 A1
20160186253 Talasaz et al. Jun 2016 A1
20160201124 Donahue et al. Jul 2016 A1
20160239602 Shendure et al. Aug 2016 A1
20160244838 Babiarz et al. Aug 2016 A1
20160257993 Fu et al. Sep 2016 A1
20160265042 Schroeder et al. Sep 2016 A1
20160289740 Fu et al. Oct 2016 A1
20160289753 Osborne et al. Oct 2016 A1
20160312276 Fu et al. Oct 2016 A1
20160319345 Gnerre et al. Nov 2016 A1
20160369333 Babiarz et al. Dec 2016 A1
20170011166 Rabinowitz et al. Jan 2017 A1
20170107576 Babiarz et al. Apr 2017 A1
20170114411 Mitchell et al. Apr 2017 A1
20170121716 Rodi et al. May 2017 A1
20170137882 Goossens et al. May 2017 A1
20170145475 Hunsley et al. May 2017 A1
20170152561 Hamamah et al. Jun 2017 A1
20170206311 Craig et al. Jul 2017 A1
20170218458 Fan et al. Aug 2017 A1
20170275689 Maguire et al. Sep 2017 A1
20170283788 Khoja et al. Oct 2017 A1
20170298427 Buis et al. Oct 2017 A1
20170314014 Green et al. Nov 2017 A1
20170342477 Jensen et al. Nov 2017 A1
20170362649 Lieberman-Aiden et al. Dec 2017 A1
20180023128 Yanai et al. Jan 2018 A1
20180025109 Rabinowitz et al. Jan 2018 A1
20180105807 Lo et al. Apr 2018 A1
20180127744 Hu et al. May 2018 A1
20180142296 Mitchell et al. May 2018 A1
20180148777 Kirkizlar et al. May 2018 A1
20180155775 Zimmermann et al. Jun 2018 A1
20180155776 Zimmermann et al. Jun 2018 A1
20180155779 Zimmermann et al. Jun 2018 A1
20180155785 Rabinowitz et al. Jun 2018 A1
20180155786 Rabinowitz et al. Jun 2018 A1
20180155792 Rabinowitz et al. Jun 2018 A1
20180171409 Rabinowitz et al. Jun 2018 A1
20180171420 Babiarz et al. Jun 2018 A1
20180173845 Sigurjonsson et al. Jun 2018 A1
20180173846 Sigurjonsson et al. Jun 2018 A1
20180187241 Selvaraj et al. Jul 2018 A1
20180201995 Rabinowitz et al. Jul 2018 A1
20180237841 Stray et al. Aug 2018 A1
20180251553 McGranahan et al. Sep 2018 A1
20180265917 Barany et al. Sep 2018 A1
20180288982 Sinha Oct 2018 A1
20180298439 Ryan et al. Oct 2018 A1
20180300448 Rabinowitz et al. Oct 2018 A1
20180303870 Golobish et al. Oct 2018 A1
20180320171 Withey Nov 2018 A1
20180320239 Babiarz et al. Nov 2018 A1
20180371531 Quake et al. Dec 2018 A1
20190010543 Babiarz et al. Jan 2019 A1
20190106737 Underhill Apr 2019 A1
20190106751 Zimmermann et al. Apr 2019 A1
20190112661 Khan et al. Apr 2019 A1
20190153521 Mitchell et al. May 2019 A1
20190153525 Mitchell et al. May 2019 A1
20190185913 Zimmermann et al. Jun 2019 A1
20190185936 Babiarz et al. Jun 2019 A1
20190194743 Ryan et al. Jun 2019 A1
20190194758 Babiarz et al. Jun 2019 A1
20190194759 Babiarz et al. Jun 2019 A1
20190203290 Rabinowitz et al. Jul 2019 A1
20190203294 Babiarz et al. Jul 2019 A1
20190211376 Quake et al. Jul 2019 A1
20190211385 Sarwar et al. Jul 2019 A1
20190211391 Rabinowitz et al. Jul 2019 A1
20190211392 Rabinowitz et al. Jul 2019 A1
20190211393 Rabinowitz et al. Jul 2019 A1
20190211399 Rabinowitz et al. Jul 2019 A1
20190211402 Babiarz et al. Jul 2019 A1
20190211406 Babiarz et al. Jul 2019 A1
20190249241 Rabinowitz et al. Aug 2019 A1
20190256894 Zimmermann et al. Aug 2019 A1
20190256906 Rabinowitz et al. Aug 2019 A1
20190256907 Ryan et al. Aug 2019 A1
20190256908 Rabinowitz et al. Aug 2019 A1
20190256909 Rabinowitz et al. Aug 2019 A1
20190256912 Rabinowitz et al. Aug 2019 A1
20190256916 Babiarz et al. Aug 2019 A1
20190256917 Babiarz et al. Aug 2019 A1
20190256919 Babiarz et al. Aug 2019 A1
20190256924 Vogelstein et al. Aug 2019 A1
20190256931 Babiarz et al. Aug 2019 A1
20190264277 Rabinowitz et al. Aug 2019 A1
20190264280 Rabinowitz et al. Aug 2019 A1
20190264288 Rabinowitz et al. Aug 2019 A1
20190271043 Babiarz et al. Sep 2019 A1
20190276888 Rabinowitz et al. Sep 2019 A1
20190284623 Rabinowitz et al. Sep 2019 A1
20190300950 Rabinowitz et al. Oct 2019 A1
20190309358 Rabinowitz et al. Oct 2019 A1
20190309359 Zimmermann et al. Oct 2019 A1
20190309365 Babiarz et al. Oct 2019 A1
20190316177 Zimmermann et al. Oct 2019 A1
20190316184 Zimmermann et al. Oct 2019 A1
20190316200 Rabinowitz et al. Oct 2019 A1
20190323076 Rabinowitz et al. Oct 2019 A1
20190360033 Stamm et al. Nov 2019 A1
20190360036 Rabinowitz et al. Nov 2019 A1
20190367972 Mitchell et al. Dec 2019 A1
20200024653 Bethke Jan 2020 A1
20200032323 Talasaz et al. Jan 2020 A1
20200032340 Mitchell Jan 2020 A1
20200109449 Stamm et al. Apr 2020 A1
20200121718 Novik et al. Apr 2020 A1
20200123612 Babiarz et al. Apr 2020 A1
20200126634 Sigurjonsson et al. Apr 2020 A1
20200140950 Babiarz et al. May 2020 A1
20200141925 Liaw et al. May 2020 A1
20200149111 Babiarz et al. May 2020 A1
20200157629 Babiarz et al. May 2020 A1
20200165678 Mitchell et al. May 2020 A1
20200172977 Rabinowitz et al. Jun 2020 A1
20200181681 Mitchell et al. Jun 2020 A1
20200181697 Rabinowitz et al. Jun 2020 A1
20200190570 Ryan et al. Jun 2020 A1
20200190573 Rabinowitz et al. Jun 2020 A1
20200190591 Rabinowitz et al. Jun 2020 A1
20200208196 Zimmermann et al. Jul 2020 A1
20200208221 Babiarz et al. Jul 2020 A1
20200224273 Rabinowitz et al. Jul 2020 A1
20200232036 Rabinowitz et al. Jul 2020 A1
20200232037 Babiarz et al. Jul 2020 A1
20200248264 Rabinowitz et al. Aug 2020 A1
20200248266 Swanton et al. Aug 2020 A1
20200206292 Wisconsin Oct 2020 A1
20200316498 Mitchell Oct 2020 A1
20200318191 Babiarz et al. Oct 2020 A1
20200347454 Babiarz et al. Nov 2020 A1
20200350034 Rabinowitz et al. Nov 2020 A1
20200362415 Rabinowitz et al. Nov 2020 A1
20200370129 Quinn et al. Nov 2020 A1
20200385809 Ramani et al. Dec 2020 A1
20200407788 Ryan et al. Dec 2020 A1
20200407798 Babiarz et al. Dec 2020 A1
20210009990 Stray et al. Jan 2021 A1
20210025005 Babiarz et al. Jan 2021 A1
20210032692 Mitchell et al. Feb 2021 A1
20210054459 Rabinowitz et al. Feb 2021 A1
20210071246 Zimmermann et al. Mar 2021 A1
20210139969 Mitchell et al. May 2021 A1
20210139983 Mitchell et al. May 2021 A1
20210139988 Mitchell et al. May 2021 A1
20210155988 Rabinowitz et al. May 2021 A1
20210189498 Babiarz et al. Jun 2021 A1
20210198733 Moshkevich et al. Jul 2021 A1
20210198742 Rabinowitz et al. Jul 2021 A1
20210198743 Babiarz et al. Jul 2021 A1
20210222230 Zimmermann et al. Jul 2021 A1
20210222240 Moshkevich et al. Jul 2021 A1
20210257048 Zimmermann et al. Aug 2021 A1
20210269879 Mitchell et al. Sep 2021 A1
20210301320 Mitchell et al. Sep 2021 A1
20210324463 Rabinowitz et al. Oct 2021 A1
20210327538 Egilsson et al. Oct 2021 A1
20210327542 Ryan et al. Oct 2021 A1
20210355536 Rabinowitz et al. Nov 2021 A1
20220025455 Zimmermann et al. Jan 2022 A1
20220025456 Rabinowitz et al. Jan 2022 A1
20220033908 Rabinowitz et al. Feb 2022 A1
20220033909 Babiarz et al. Feb 2022 A1
20220042103 Rabinowitz et al. Feb 2022 A1
20220056509 Zimmermann Feb 2022 A1
20220056534 Rivers Feb 2022 A1
20220073978 Rabinowitz et al. Mar 2022 A1
20220073979 Rabinowitz et al. Mar 2022 A1
20220098667 Rabinowitz et al. Mar 2022 A1
20220139495 Rabinowitz et al. May 2022 A1
20220145391 Mitchell et al. May 2022 A1
20220154249 Zimmermann et al. May 2022 A1
20220154290 Babiarz et al. May 2022 A1
20220195526 Rabinowitz et al. Jun 2022 A1
20220213561 Babiarz et al. Jul 2022 A1
20220251654 Hafez et al. Aug 2022 A1
20220267849 Mitchell et al. Aug 2022 A1
20220307086 Babiarz et al. Sep 2022 A1
20220356522 Mitchell et al. Nov 2022 A1
20220356526 Babiarz et al. Nov 2022 A1
20220356530 Sharma Nov 2022 A1
20220403461 Kirkizlar et al. Dec 2022 A1
20220411875 Rabinowitz et al. Dec 2022 A1
20230054494 Rabinowitz et al. Feb 2023 A1
20230053752 Rabinowitz et al. Mar 2023 A1
20230060579 Bethke et al. Mar 2023 A1
20230193387 Rabinowitz Jun 2023 A1
20230203573 Swenerton et al. Jun 2023 A1
20230212693 Rabinowitz et al. Jul 2023 A1
20230242998 Babiarz et al. Aug 2023 A1
20230332221 Zimmermann et al. Oct 2023 A1
20230343411 Rabinowitz et al. Oct 2023 A1
Foreign Referenced Citations (307)
Number Date Country
112017023232 Aug 2018 BR
2875281 Dec 2013 CA
1650032 Aug 2005 CN
1674028 Sep 2005 CN
101675169 Mar 2010 CN
102892901 Jan 2013 CN
104736722 Jun 2015 CN
105229175 Jan 2016 CN
107365769 Nov 2017 CN
107849604 Mar 2018 CN
109661476 Apr 2019 CN
201792389 May 2018 EA
0270017 Jun 1988 EP
1325963 Jul 2003 EP
1524321 Apr 2005 EP
1325963 Sep 2006 EP
1524321 Jul 2009 EP
2163622 Mar 2010 EP
2128169 Dec 2010 EP
2551356 Jan 2013 EP
2653562 Oct 2013 EP
2902500 Aug 2015 EP
3026124 Jun 2016 EP
2315849 Nov 2017 EP
3285193 Feb 2018 EP
2877594 Dec 2019 EP
3187597 Jun 2020 EP
3134541 Aug 2020 EP
3760730 Jan 2021 EP
3760731 Jan 2021 EP
3760732 Jan 2021 EP
3824470 May 2021 EP
3443119 Feb 2022 EP
2488358 Aug 2012 GB
2965699 Aug 1999 JP
2002-530121 Sep 2002 JP
2002-300894 Oct 2002 JP
2003521252 Jul 2003 JP
2004502466 Jan 2004 JP
2004121087 Apr 2004 JP
2004533243 Nov 2004 JP
2005514956 May 2005 JP
2005160470 Jun 2005 JP
2006-254912 Sep 2006 JP
2008-263974 Nov 2008 JP
2008271980 Nov 2008 JP
2010-509922 Apr 2010 JP
2011508662 Mar 2011 JP
2011516069 May 2011 JP
2012-085556 May 2012 JP
2013509883 Mar 2013 JP
2015-535681 Dec 2015 JP
2016502849 Feb 2016 JP
2290078 Dec 2006 RU
9501796 Jan 1995 WO
9623067 Aug 1996 WO
1996036736 Nov 1996 WO
9839474 Sep 1998 WO
9844151 Oct 1998 WO
9937773 Jul 1999 WO
0018957 Apr 2000 WO
2001007640 Feb 2001 WO
0134844 May 2001 WO
0157269 Aug 2001 WO
2001079851 Oct 2001 WO
200190419 Nov 2001 WO
2002004672 Jan 2002 WO
0244411 Jun 2002 WO
0244411 Jun 2002 WO
2002055985 Jul 2002 WO
02070751 Sep 2002 WO
02070751 Sep 2002 WO
2002076377 Oct 2002 WO
02090505 Nov 2002 WO
03000919 Jan 2003 WO
03018757 Mar 2003 WO
03020974 Mar 2003 WO
2003031646 Apr 2003 WO
2003050532 Jun 2003 WO
2003062441 Jul 2003 WO
2003102595 Dec 2003 WO
2003106623 Dec 2003 WO
2004051218 Jun 2004 WO
2004069849 Aug 2004 WO
2004070005 Aug 2004 WO
2004070007 Aug 2004 WO
2004078999 Sep 2004 WO
2004081183 Sep 2004 WO
2004087863 Oct 2004 WO
2005003375 Jan 2005 WO
2005021793 Mar 2005 WO
2005023091 Mar 2005 WO
2005030999 Apr 2005 WO
2005035725 Apr 2005 WO
2005039389 May 2005 WO
2005100401 Oct 2005 WO
2005123779 Dec 2005 WO
2007145612 Jun 2006 WO
2006110855 Oct 2006 WO
2006128192 Nov 2006 WO
2007011903 Jan 2007 WO
2007052006 May 2007 WO
2007057647 May 2007 WO
2007062164 May 2007 WO
2007073171 Jun 2007 WO
2007070280 Jun 2007 WO
2007070482 Jun 2007 WO
2007075836 Jul 2007 WO
2007092473 Aug 2007 WO
2007086935 Aug 2007 WO
2007117256 Oct 2007 WO
2007117039 Oct 2007 WO
2007132167 Nov 2007 WO
2007147073 Dec 2007 WO
2007147076 Dec 2007 WO
2007140417 Dec 2007 WO
2007147074 Dec 2007 WO
2007147079 Dec 2007 WO
2008024473 Feb 2008 WO
2008048931 Apr 2008 WO
2008061213 May 2008 WO
2008051928 May 2008 WO
2008056937 May 2008 WO
2008059578 May 2008 WO
2008079374 Jul 2008 WO
2008081451 Jul 2008 WO
2008084405 Jul 2008 WO
2008115427 Sep 2008 WO
2008115497 Sep 2008 WO
2008118988 Oct 2008 WO
2008135837 Nov 2008 WO
2008157264 Dec 2008 WO
2009009769 Jan 2009 WO
2009013492 Jan 2009 WO
2009013496 Jan 2009 WO
2009019215 Feb 2009 WO
2009019455 Feb 2009 WO
2009032779 Mar 2009 WO
2009036525 Mar 2009 WO
2009030100 Mar 2009 WO
2009032781 Mar 2009 WO
2009033178 Mar 2009 WO
2009049889 Apr 2009 WO
2009017784 May 2009 WO
2009064897 May 2009 WO
2009091934 Jul 2009 WO
2009092035 Jul 2009 WO
2009105531 Aug 2009 WO
2009099602 Aug 2009 WO
2009100029 Aug 2009 WO
2009117122 Sep 2009 WO
2009120808 Oct 2009 WO
2009145828 Dec 2009 WO
2009146335 Dec 2009 WO
2010014920 Feb 2010 WO
2010017214 Feb 2010 WO
2010033639 Mar 2010 WO
2010033652 Mar 2010 WO
2010033578 Mar 2010 WO
2010042831 Apr 2010 WO
2010045617 Apr 2010 WO
2010075459 Jul 2010 WO
2010088288 Aug 2010 WO
2010115016 Oct 2010 WO
2010115154 Oct 2010 WO
2010118016 Oct 2010 WO
2010127186 Nov 2010 WO
2011015944 Feb 2011 WO
2011023078 Mar 2011 WO
2011032078 Mar 2011 WO
2011041485 Apr 2011 WO
2011051283 May 2011 WO
2011057061 May 2011 WO
2011057094 May 2011 WO
2011090556 Jul 2011 WO
2011087760 Jul 2011 WO
2011094646 Aug 2011 WO
2011102998 Aug 2011 WO
2011118603 Sep 2011 WO
2011109440 Sep 2011 WO
2011118603 Sep 2011 WO
2011142836 Nov 2011 WO
2011140433 Nov 2011 WO
2011146632 Nov 2011 WO
2012019200 Feb 2012 WO
2012028746 Mar 2012 WO
2012042374 Apr 2012 WO
2012058488 May 2012 WO
2012-083189 Jun 2012 WO
201283250 Jun 2012 WO
2012088456 Jun 2012 WO
20120071621 Jun 2012 WO
2012092426 Jul 2012 WO
2012108920 Aug 2012 WO
2012122374 Sep 2012 WO
2012142531 Oct 2012 WO
2007149791 Dec 2012 WO
2013030577 Mar 2013 WO
2013045432 Apr 2013 WO
2013049892 Apr 2013 WO
2013052557 Apr 2013 WO
2013078470 May 2013 WO
2013086464 Jun 2013 WO
2013123220 Aug 2013 WO
2013138510 Sep 2013 WO
2013138510 Sep 2013 WO
20130130848 Sep 2013 WO
2013159035 Oct 2013 WO
2013169339 Nov 2013 WO
2013177220 Nov 2013 WO
2013181651 Dec 2013 WO
2013190441 Dec 2013 WO
2014004726 Jan 2014 WO
2014014497 Jan 2014 WO
20140018080 Jan 2014 WO
2014026277 Feb 2014 WO
2014035986 Mar 2014 WO
2014039556 Mar 2014 WO
2014099919 Jun 2014 WO
2014122288 Aug 2014 WO
20141424290 Aug 2014 WO
2014118334 Aug 2014 WO
2014145078 Sep 2014 WO
2014145232 Sep 2014 WO
2014149134 Sep 2014 WO
2014150300 Sep 2014 WO
2014151117 Sep 2014 WO
2014143989 Sep 2014 WO
2014194113 Dec 2014 WO
2015035177 Mar 2015 WO
2015134552 Mar 2015 WO
2015134552 Mar 2015 WO
2015048535 Apr 2015 WO
2015070086 May 2015 WO
2015069933 May 2015 WO
2015100427 Jul 2015 WO
2015138997 Sep 2015 WO
2015148494 Oct 2015 WO
2015164432 Oct 2015 WO
2015169947 Nov 2015 WO
2015178978 Nov 2015 WO
2016009059 Jan 2016 WO
2016001411 Jan 2016 WO
2016009224 Jan 2016 WO
2016028316 Feb 2016 WO
2016063122 Apr 2016 WO
2016065295 Apr 2016 WO
2016077313 May 2016 WO
2016123698 Aug 2016 WO
2016138080 Sep 2016 WO
2016176662 Nov 2016 WO
2016183106 Nov 2016 WO
2016193490 Dec 2016 WO
2016192956 Dec 2016 WO
2017011329 Jan 2017 WO
2017-045654 Mar 2017 WO
2017058784 Apr 2017 WO
2017091865 Jun 2017 WO
2017176852 Oct 2017 WO
2017181146 Oct 2017 WO
2017181202 Oct 2017 WO
2017190106 Nov 2017 WO
2017205540 Nov 2017 WO
2018009723 Jan 2018 WO
2018083467 May 2018 WO
2018085603 May 2018 WO
2018085597 May 2018 WO
2018106798 Jun 2018 WO
2018119422 Jun 2018 WO
2018136562 Jul 2018 WO
2018156418 Aug 2018 WO
2018237081 Dec 2018 WO
2018237078 Dec 2018 WO
2019006561 Jan 2019 WO
2019008408 Jan 2019 WO
2019046817 Mar 2019 WO
2019053243 Mar 2019 WO
2019118926 Jun 2019 WO
2019109053 Jun 2019 WO
2019140298 Jul 2019 WO
2019161244 Aug 2019 WO
2019200228 Oct 2019 WO
2019241349 Dec 2019 WO
2020010255 Jan 2020 WO
2020018522 Jan 2020 WO
2020041449 Feb 2020 WO
2020076957 Apr 2020 WO
2020106987 May 2020 WO
2020104670 May 2020 WO
2020131699 Jun 2020 WO
2020131955 Jun 2020 WO
2020206269 Oct 2020 WO
2020214547 Oct 2020 WO
2020206290 Oct 2020 WO
2020247263 Dec 2020 WO
2021055968 Mar 2021 WO
2007100911 Sep 2021 WO
2021243045 Dec 2021 WO
2022015676 Jan 2022 WO
2022182878 Sep 2022 WO
2022197864 Sep 2022 WO
2023014597 Feb 2023 WO
2023034090 Mar 2023 WO
2023133131 Jul 2023 WO
2011130751 Oct 2023 WO
2011146942 Nov 2023 WO
2011153254 Dec 2023 WO
Non-Patent Literature Citations (1332)
Entry
US 8,501,409 B2, 08/2013, Simen et al. (withdrawn)
Vanessa Garćia Moreira et al, “Cell-Free DNA as a Noninvasive Acute Rejection Marker in Renal Transplantation” Clinical Chemistry, vol. 55, Issue 11, Nov. 1, 2009, pp. 1958-1966 (Year: 2009).
Olerup, O. et al. Tissue Antigens 1992: 39: 225-235. (Year: 1992).
Snyder, T.M. et al. “Universal noninvasive detection of solid organ transplant rejection” PNAS, Apr. 12, 2011, vol. 198, No. 15, p. 6229-6234. (Year: 2011).
Nakamura, N. et al. “Ex Vivo Liver Perfusion with Arterial Blood from a Pig with Ischemic Liver Failure” Artificial Organs 23(2):153-160. (Year: 1999).
Nui, A. et al “The Functional Integrity of a Normothermic Perfusion System UsingArtificial Blood in Pig Liver” Journal of Surgical Research 131, 189-198 (Year: 2006).
Stone, J.P. et al. “Ex Vivo Normothermic Perfusion Induces Donor-Derived Leukocyte Mobilization and Removal Prior to Renal Transplantation” Kidney International Reports (Aug. 6, 2016) 1, 230-239 (Year: 2016).
Norton, S.E. et al. “A stabilizing reagent prevents cell-free DNA contamination by cellular DNA in plasma during blood sample storage and shipping as determined by digital PCR”. Clinical Biochemistry 46 (2013) 1561-1565 (Year: 2013).
Amador Goncalves-Primo et al. “Investigation of Apoptosis-Related Gene Expression Levels in Preimplantation Biopsies as Predictors of Delayed Kidney Graft Function” Transplantation & vol. 97, No. 12, Jun. 27, (Year: 2014).
Kanou, T. et al. “Cell-Free DNA in Human Ex-Vivo Lung Perfusate as a Potential Biomarker to Predict the Risk of Primary Graft Dysfunction (PGD) in Lung Transplantation” The Journal of Heart and Lung Transplantation, vol. 36, Iss. 4, Supplement, Abstract No. 187 (Apr. 1, 2017) (Year: 2017).
Markus Selzner, et al., “Normothermic Ex Vivo Liver Perfusion Using Steen Solution as Perfusate for Human Liver Transplantation: First North American Results” Liver Transplantation, vol. 22, Issue11, Nov. 2016 (First published: Jun. 24, 2016) (Year: 2016).
S. H. Keshavjee, et al. “The role of dextran 40 and potassium in extended hypothermic lung preservation for transplantation” The Journal of Thoracic and Cardiovascular Surgery, vol. 103, No. 2, Feb. 1992. (Year: 1992).
Dirk Van Raemdonck et al., “Ex-vivo lung perfusion” Transplant International, vol. 28, Issue 6, Special Issue: Focus Issue: Machine Perfusion, Jun. 2015, pp. 643-656 (First published: Mar. 15, 2014) (Year: 2014).
F. Valenza, et al. “The Consumption of Glucose During Ex Vivo Lung Perfusion Correlates With Lung Edema” Transplantation Proceedings, vol. 43, Issue 4, May 2011, pp. 993-996 (Year: 2011).
J.P. Stone, et al. “Altered Immunogenicity of Donor Lungs via Removal of Passenger Leukocytes Using Ex Vivo Lung Perfusion” American Journal of Transplantation, vol. 16, Issue 1, Jan. 2016, pp. 33-43 (Year: 2016).
International Search Report and Written Opinion for Application No. PCT/US2018/065845 mailed Mar. 8, 2019.
International Preliminary Report on Patentability for Application No. PCT/US2018/065845 mailed Jun. 25, 2020.
Crespo et al., Pre-transplant Donor-Specific T-cell Alloreactivity Is Strongly Associated With Early Acute Cellular Rejection in Kidney Transplant Recipients Not Receiving T-cell Depleting Induction Therapy. PLoS One. Feb. 17, 2015;10(2):e0117618. doi: 10.1371/journal.pone.0117618. eCollection 2015.
Lehmann-Werman et al., Identification of Tissue-Specific Cell Death Using Methylation Patterns of Circulating DNA. Proc Natl Acad Sci U S A. Mar. 29, 2016;113(13):E1826-34. doi: 10.1073/pnas.1519286113. Epub Mar. 14, 2016.
Partpart-Li et al., The Effect of Preservative and Temperature on the Analysis of Circulating Tumor DNA. Clin Cancer Res. May 15, 2017;23(10):2471-2477. doi: 10.1158/1078-0432.CCR-16-1691. Epub Nov. 8, 2016.
Sairafi et al., Donor Cell Composition and Reactivity Predict Risk of Acute Graft-versus-Host Disease After Allogeneic Hematopoietic Stem Cell Transplantation. J Immunol Res. 2016;2016:5601204. doi: 10.1155/2016/5601204. Epub Nov. 14, 2016.
Snyder et al., Cell-free DNA Comprises an In Vivo Nucleosome Footprint That Informs Its Tissues-Of-Origin. Cell. Jan. 14, 2016;164(1-2):57-68. doi: 10.1016/j.cell.2015.11.050.
Verhoeven et al., Biomarkers to Assess Graft Quality During Conventional and Machine Preservation in Liver Transplantation. J Hepatol. Sep. 2014;61(3):672-84. doi: 10.1016/j.jhep.2014.04.031. Epub May 4, 2014.
Ahmadian, A. et al., “Analysis of the p53 Tumor Suppressor Gene by Pyrosequencing”, Bio Techniques, vol. 28, Jan. 2000, 140-147.
Birkenkamp-Demtroder, et al., “Longitudinal assessment of multiplex patient-specific ctDNA biomarkers in bladder cancer for diagnosis, surveillance and recurrence”, Annals of Oncology, Oxford University Press NLD, vol. 29, No. Supplement 8, 2018, viii26.
Bolotin, D. A. et al., “MiXCR: software for comprehensive adaptive immunity profiling”, Nature, vol. 12, No. 5, May 2015, 380-381.
Brochet, X. et al., “IMGT/V-QUEST: the highly customized and integrated system for IG and TR standardized V-J and V-D-J sequence analysis”, Nucleic Acids Research, vol. 36, May 23, 2008, W503-W508.
Bunnapradist, S. et al., “Using both the fraction and Quantity of Donor-Derived Cell-free DNA to Detect Kidney Allograft Rejection”, JASN, vol. 32, 2021, 2439-2441.
Burnham, P. et al., “Single-stranded DNA library preparation uncovers the origin and diversity of ultrashort cell-free DNA in plasma”, Scientific Reports, vol. 6, No. 27859, Jun. 14, 2016, 9 pages.
Cawkwell, L. et al., “Rapid detection of allele loss in colorectal tumours using microsatellites and fluorescent DNA technology”, Br. J. Cancer, vol. 67, 1993, 1262-1267.
Chen, et al., “Non-invasive prenatal diagnosis using fetal DNA in maternal plasma: a preliminary study for identification of paternally-inherited alleles using single nucleotide polymorphisms”, BMJ Open, 5(7), 2015, 1-8.
Chothia, C. et al., “Canonical structures for the hypervariable regions of immunoglobulins”, Journal of Molecular Biology, vol. 196, No. 4, Aug. 20, 1987, 901-917.
Chun, et al., “Dual priming oligonucleotide system for the multiplex detection of respiratory viruses and SNP genotyping of CYP2C19 gene”, Nucleic Acids Research, vol. 35, No. 6, 2007, 1-6.
Costa, J.-M et al., “Fetal RHD genotyping in maternal serum during the first trimester of pregnancy”, British Journal of Haematology, vol. 119, 2002, 255-260.
Croft, Jr., Daniel et al., “Performance of Whole-Genome Amplified DNA Isolated from Serum and Plasma on High-Density Single Nucleotide Polymorphism Arrays”, Journal of Molecular Diagnostics, 10(3), 2008, 249-257.
Daniels, G. et al., “Fetal blood group genotyping from DNA from maternal plasma: an important advance in the management and prevention of haemolytic disease of the fetus and newborn”, Vox Sanguinis, vol. 87, 2004, 223-232.
Deusen, et al., “Comprehensive Detection of Driver Mutations in Acute Myeloid Leukemia Including Internal Tandem Duplications with Anchored Multiplex PCR and Next-Generation Sequencing”, Blood, vol. 128, No. 22, 2016, 5251.
Diaz, et al., “Liquid Biopsies: Genotyping Circulating Tumor DNA”, Journal of Clinical Oncology, vol. 32, No. 6, 2014, 579-586.
Ehlayel, A. et al., “Emerging monitoring technologies in kidney transplantation”, Pediatric Nephrology, vol. 36, 2021, 3077-3087.
Findlay, I. et al., “Allelic drop-out and preferential amplification in single cells and human blastomeres: implications for preimplantation diagnosis of sex and cystic fibrosis”, Molecular Human Reproduction, vol. 1, 1995, 1609-1618.
Fouquet, C. et al., “Rapid and Sensitive p53 Alteration Analysis in Biopsies from Lung Cancer Patients Using a Functional Assay and a Universal Oligonudeotide Array: A Prospective Study”, Clinical Cancer Research, vol. 10, May 15, 2004, 3479-3489.
Fournie, et al., “Plasma DNA as a Marker of Cancerous Cell Death. Investigations in Patients Suffering From Lung Cancer and in Nude Mice Bearing Human Tumours”, Cancer Letters, vol. 91, No. 2, May 8, 1995, 221-227.
Fredriksson, et al., “Multiplex amplification of all coding sequences within 10 cancer genes by Gene-Collector”, Nucleic Acids Research, 2007, vol. 35, No. 7 e47, 1-6.
Fredriksson, M et al., “Assessing Hematopoietic Chimerism After Allogeneic Stem Cell Transplantation by Multiplexed SNP Genotyping Using Microarrays and Quantitive Analysis of SNP Alleles”, Leukemia, vol. 18, Issue 2, Dec. 4, 2003, 255-266.
Freeman, Jennifer L. et al., “Copy Number Variation: New Insights in Genome Diversity”, Genome Research, 16, 2006, 949-961.
Frohman, M A. et al., “On Beyond Classic RACE (Rapid Amplification of cDNA Ends)”, Genome Research, vol. 4, 1994, S40-S58.
Frost, Mackenzie S et al., “Differential Effects of Chronic Pulsatile Versus Chronic Constant Maternal Hyperglycemia on Fetal Pancreatic B-Cells”, Journal of Pregnancy, 2012,, Article ID 812094, 2012, 8.
Fu, G. K. et al., “Counting individual DNA molecules by the stochastic attachment of diverse labels”, PNAS, vol. 108, No. 22, May 31, 2011, 9026-9031.
Garcia Moreira, V. et al., “Cell-Free DNA as a Noninvasive Acute Rejection Marker in Renal Transplantation”, Clinical Chemistry, vol. 55, No. 11, 2009, 1958-1966.
Glaab, W. E. et al., “A novel assay for allelic discrimination that combines the fluorogenic 5′ nuclease polymerase chain reaction (TaqMan) and mismatch amplification mutation assay”, Mutation Research, vol. 430, 1999, 12 pgs.
Goncalves-Primo, A. et al., “Investigation of Apoptosis-Related Gene Expression Levels in Preimplantation Biopsies as Predictors of Delayed Kidney Graft Function”, Transplantation, vol. 97, No. 12, Jun. 27, 2014.
Grenda, R. “Torque teno (TTV) viral load as a biomarker of immunosuppressive strength after kidney transplantation in children”, Pediatric Nephrology, vol. 36, May 27, 2020, 3 pages.
Gusella, J. et al., “Precise localization of human B-globin gene complex on chromosome 11*”, Proc. Natl. Acad. Sci USA, vol. 76, No. 10, Oct. 1979, 5239-5243.
Hainer & Fazzio, “High-Resolution Chromatin Profiling Using Cut&Run”, Current Protocols in Molecular Biology, 2019, 1-22.
Hara, Eiji et al., “Subtractive eDNA cloning using oligo(dT)3o-latex and PCR: isolation of eDNA clones specific to undifferentiated human embryonal carcinoma cells”, Nucleic Acids Research, 19(25), 1991, 7097-7104.
Hiendleder, et al., “Functional genomics: tools for improving farm animal health and welfare”, Rev. Sci. Tech. Off. Int. Epiz., 24 (1), 2005, 354-377.
Hug, H. et al., “Measurement of the number of molecules of a single mRNA species in a complex mRNA preparation”, J. Theor. Biol., vol. 221, 2003, 615-624.
Illumina, “HumanOmni1-Quad BeadChip”, Illumina DNA Analysis, Pub. No. 370-21009-007, 2009, 1 page.
Illumina, “HumanOmni2.5-8 BeadChips: Next-Generation GWAS Content for Genotyping and CNV Analysis”, Data Sheet: DNA Analysis, Pub. No. 370-2011-008, 2011, 1 page.
Illumina, “Illumina Adapter Sequences”, Published by Illumina, 2018, 1-45.
Jordens, et al., “Amplification with molecular beacon primers and reverse line blotting for the detection and typing of human papillomaviruses”, Journal of Virological Methods, vol. 89, 2000, 29-37.
Kaboev, et al., “PCR hot start using primers with the structure of molecular beacons (hairpin-like structure)”, Nucleic Acids Research, vol. 28, 2000, 1-2.
Kanou, et al., “Cell-free DNA in human ex vivo lung perfusate as a potential biomarker to predict the risk of primary graft dysfunction in lung transplantation”, The Journal of Heart and Lung Transplantation, vol. 36, No. 45, 2017, S187.
Keshavjee, S. H. et al., “The role of dextran 40 and potassium in extended hypothermic lung preservation for transplantation”, The Journal of Thoracic and Cardiovascular Surgery, vol. 103, No. 2, 1992.
Kittler, R. et al., “A Whole Genome Amplification Method to Generate Long Fragments from Low Quantities of Genomic DNA”, Analytical Biochemistry, vol. 300, 2002, 237-244.
Kivioja, T. et al., “Counting absolute No. of molecules using unique molecular identifiers”, Nature Proceedings, Apr. 14, 2011, 18 pgs.
Kivioja, T. et al., “Counting absolute Nos. of molecules using unique molecular identifiers”, Nature Methods, Advance Online Publication, Nov. 20, 2011, 1-5.
Kulifaj, D. et al., “Development of a standardized real time PCR for Torque teno viruses (TTV) viral load detection and quantification: A new tool for immune monitoring”, Journal of Clinical Virology, vol. 105, 2018, 118-127.
Lajoie, B. R. et al., “The Hitchhiker's Guide to Hi-C Analysis: Practical guidelines”, Methods: Author manuscript, vol. 72, Jan. 2015, 65-75.
Landegren, U. et al., “Reading Bits of Genetic Information: Methods for Single-Nucleotide Polymorphism Analysis”, Genome Research, vol. 8, No. 8, 769-776, 1997.
Li, Ying et al., “Ready detection of donor-specific single-nucleotide polymorphisms in the urine of renal transplant recipients by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry”, Clin Chem, Oct. 2005, vol. 51, Issue.10,pp. 1903-1904, Oct. 1, 2005, 1903-1904.
Lo, Y.M. D. et al., “Prenatal Diagnosis of Fetal RhD Status by Molecular Analysis of Maternal Plasma”, The New England Journal of Medicine, vol. 339, No. 24, 1998, 1734-1738.
McBride, D. et al., “Use of Cancer-Specific Genomic Rearrangements to Quantify Disease Burden in Plasma from Patients with Solid Tumors”, Genes, Chromosomes & Cancer, vol. 49, Aug. 19, 2010, 1062-1069.
Miner, B. E. et al., “Molecular barcodes detect redundancy and contamination in hairpin-bisulfite PCR”, Nucleic Acids Research, vol. 32, No. 17, Sep. 30, 2004, 1-4.
Nakamura, N. et al., “Ex Vivo Liver Perfusion with Arterial Blood from a Pig with Ischemic Liver Failure”, Artificial Organs, vol. 23, No. 2, 1999, 153-160.
Namlos, H.M. et al., “Use of liquid biopsies to monitor disease progression in a sarcoma patient: a case report”, BMC Cancer, vol. 17, No. 1, 2017, 2-3.
NCBI, “dbSNP record for rs2384571”, Retrieved from the Internet: <URL: www.ncbi.nlm.nih.gov/snp/?term=rs2384571>, 2019, 2 pgs.
NCBI, “dbSNP record for rs3731877”, Retrieved from the Internet: <URL: www.ncbi.nlm.nih.gov/snp/?term=rs3731877>, 2019, 2 pgs.
Nelson, C. M. et al., “Whole genome transcription profiling of Anaplasma phagocytohilum in human and tick host cells by tiling array analysis”, BMC Genomics, vol. 9, No. 364, Jul. 31, 2008, 16 pgs.
Nicolaides, K. et al., “Noninvasive Prenatal Testing for Fetal Trisomies in a Routinely Screened First-Trimester Population”, American Journal of Obstetrics (article in press), 207, 2012, 1.e1-1.e6.
Nicolaides, K.H et al., “Validation of Targeted Sequencing of Single-Nucleotide Polymorphisms for Non-Invasive Prenatal Detection of Aneuploidy of Chromosomes 13, 18, 21, X, and Y”, Prenatal Diagnosis, 33, 2013, 575-579.
Nilsson, et al., “Analyzing genes using closing and replicating circles”, Trends in Biotechnology, 24, 2006, 83-88.
Norton, S. E. et al., “A stabilizing reagent prevents cell-free DNA contamination by cellular DNA in plasma during blood sample storage and shipping as determined by digital PCR”, Clin Biochem., vol. 46, No. 15, Oct. 2013, 1561-1565.
Nui, A. et al., “The Functional Integrity of a Normothermic Perfusion System Using Artificial Blood in Pig Liver”, Journal of Surgical Research, Vo. 131, 2006, 189-198.
Ohira, T. et al., “Tumor vol. determines the feasibility of cell-free DNA sequencing for mutation detection in non-small cell lung cancer”, Cancer Science, vol. 107, 2016, 1660-1666.
Ohya, K. et al., “Detection of the CTG Repeat Expansion in Congenital Myotonic Dystrophy”, Jpn J. Human Genet, vol. 42, 1997, 169-180.
Olerup, O. et al., “HLA-DR typing by PCR amplification with sequence-specific primers (PCR-SSP) in 2 hours: an alternative to serological DR typing in clinical practice including donor-recipient matching in cadaveric transplantation”, Tissue Antigens, vol. 39, No. 5, May 1992, 225-235.
Ozawa, Makiko et al., “Two Families with Fukuyama Congenital Muscular Dystrophy that Underwent In Utero Diagnosis Based on Polymorphism Analysis”, Clinical Muscular Dystrophy: Research in Immunology and Genetic Counseling—FY 1994 Research Report, (including text in Japanese), 1994, 8.
Peters, David P. et al., “Noninvasive Prenatal Diagnosis of a Fetal Microdeletion Syndrome”, New England Journal of Medicine, 365(19), 2011, 1847-1848.
Philip, J. et al., “Late First-Trimester Invasive Prenatal Diagnosis: Results of an International Randomized Trial”, American College of Obstetricians and Gynecologists, vol. 103, No. 6, Jun. 2004, 1164-1173.
Pinard, et al., “Assessment of Whole Genome Amplification-induced Bias Through High-throughput, Massively Parallel Whole Genome Sequencing”, BMC Genomics, vol. 7:216, Aug. 23, 2006, 1-21.
Pourmand, et al., “Multiplex Pyrosequencing”, Nucleic Acid Research, vol. 30, No. 7, Apr. 1, 2002, 1-5.
Puszyk, William M. et al., “Noninvasive Prenatal Diagnosis of Aneuploidy Using Cell-free Nucleic Acids in Maternal Blood: Promises and Unanswered Questions”, Prenatal Diagnosis, vol. 28, No. 1, Nov. 16, 2007, 1-6.
Qiagen, QIAamp DNA Mini Kit and QIAamp DNA Blood Mini Kit Handbook, Feb. 2003 (“Qiagen (2003)”), 2003, 68 pages.
Raemdonck, Dirk Van et al., “Ex-vivo lung perfusion”, Transplant International, vol. 28, Issue 6, Special Issue: Focus Issue: Machine Perfusion, 2014, 643-656.
Rechitsky, S. et al., “Allele Dropout in Polar Bodies and Blastomeres”, Journal of Assisted Reproduction and Genetics, vol. 15, No. 5, 1998, 253-257.
Rechitsky, Svetlana et al., “Preimplantation Genetic Diagnosis with HLA Matching”, Reproductive Bio Medicine Online, 9, 2, 2004, 210-221.
Ryan, A. et al., “Informatics-Based, Highly Accurate, Noninvasive Prenatal Paternity Testing”, Genetics in Medicine (advance online publication), 2012, 5 pgs.
Ryan, B. M. et al., “A prospective study of circulating mutant KRAS2 in the serum of patients with colorectal neoplasia: strong prognostic indicator in postoperative follow up”, Gut, vol. 52, 2003, 101-108.
Schutz, E. et al., “Graft-derived cell-free DNA, a noninvasive early rejection and graft damage marker in liver transplantation: A prospective, observational, multicenter cohort study”, PLOS Medicine, vol. 14, No. 4, Apr. 25, 2017, 19 pgs.
Selzner, Markus et al., “Normothermic Ex Vivo Liver Perfusion Using Steen Solution as Perfusate for Human Liver Transplantation: First North American Results”, Liver Transplantation, vol. 22, Issue 11, 2016.
Sethi, Himanshu et al., “Analytical validation of the Signatera (TM) RUO assay, a highly sensitive patient-specific multiplex PCR NGS-based noninvasive cancer recurrence detection and therapy monitoring assay”, Cancer Research, vol. 78, No. 13, 2018, 4542.
Sigdel, T. et al., “Plasma Donor-Derived Cell-Free DNA Quantification by massively multiplex PCR Distinguishes Kidney Transplant Acute Rejection”, Transplantation, vol. 102, No. 7S, Jul. 2018, S178-S179.
Snyder, T. M. et al., “Universal noninvasive detection of solid organ transplant rejection”, PNAS, vol. 108, No. 15, Apr. 12, 2011, 6229-6234.
Solexa, “Application Note: DNA Sequencing”, 2006, pp. 1-2.
Stone, J. P. et al., “Ex Vivo Normothermic Perfusion Induces Donor-Derived Leukocyte Mobilization and Removal Prior to Renal Transplantation”, Kidney Int Rep., vol. 1, No. 4, Aug. 6, 2016, 230-239.
Tie, et al., “Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer”, Annals of Oncology, vol. 26, No. 8, 2015, 1715-1722.
Toth, T. et al., “Prenatal Detection of Trisomy 13 From Amniotic Fluid by Quantitative Fluorescent Polymerase Chain Reaction”, Prenatal Diagnosis, vol. 18, 1998, 669-674.
Tungwiwat, et al., “Non-invasive fetal sex determination using a conventional nested PCR analysis of fetal DNA in maternal plasma”, Clinica Chimica Acta, vol. 334, No. 1-2, 2003, 173-177.
Ventura-Aguiar, P. et al., “Donor-derived Cell-free DNA Shows High Sensitivity for the Diagnosis of Pancreas Graft Rejection in Simultaneous Pancreas-Kidney Transplantation”, Transplantation, vol. 00, No. 00, 2022, 8 pages.
Vlaminck, I. D. et al., “Circulating Cell-Free DNA Enables Noninvasive Diagnosis of Heart Transplant Rejection”, Sci Transl Med., vol. 6, No. 241, Jun. 18, 2014, 26 pages.
Volckmar, et al., “A field guide for cancer diagnostics using cell-free DNA: From principles to practice and clinical applications”, Genes Chromosomes Cancer, 2018, 123-139.
Wagner, F. F. et al., “RHD gene deletion occurred in the Rhesus box”, Blood, vol. 95, No. 12, 2000, 3662-3668.
Wang, et al., “Molecular inversion probes: a novel microarray technology and its application in cancer research”, Cancer Genetics, 205, 2012, 341-355.
Wangkumhang, P. et al., “WASP: a Web-based Allele-Specific PCR assay designing tool for detecting SNPs and mutations”, BMC Genomics, vol. 8, No. 275, Aug. 14, 2007, 9 pgs.
Whitlam, J. B. et al., “Diagnostic application of kidney allograft-derived absolute cell-free DNA levels during transplant dysfunction”, Am J Transplant, vol. 19, 2019, 1037-1049.
Ye, et al., “Primer-BLAST: a tool to design target-specific primers for polymerase chain reaction”, BMC Bioinformatics, 13:134, 2012, 11 pages.
Benesova, et al., “Mutation-based detection and monitoring of cell-free tumor DNA in peripheral blood of cancer patients”, Analytical Biochemistry, vol. 433, 2013, 227-234.
Diehl, et al., “Detection and quantification of mutations in the plasma of patients with colorectal tumors”, Proceedings of the National Academy of Sciences, vol. 102, 2005, 16368-16373.
Ku, et al., “Exome versus transcriptome sequencing in identifying coding region variants”, Expert Review of Molecular Diagnostics, vol. 12, 2012, 241-251.
Marusyk, et al., “Causes and consequences”, Biochimica et Biophysica Acta, vol. 1805, 2010, 105-117.
Birkenkamp-Demtroder, et al., “Longitudinal assessment of multiplex patient-specific ctDNA biomarkers in bladder cancer for diagnosis, surveillance and recurrence”, Annals of Oncology.
Oxford University Press NLD, vol. 29, No. Supplement 8, 2018, viii26.
18820195.8, “Extended European Search Report”, mailed Jan. 27, 2021, 9 pages.
18821381.3, Extended European Search Report, mailed Feb. 15, 2021, 9 pages.
Abbosh, et al., “Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution”, Nature, 2017, 446-453.
Adamek, Martina et al., “A fast and simple method for detecting and quantifying donor-derived cell-free DNA in sera of solid organ transplant recipients as a biomarker for graft function”, Clinical Chemistry and Laboratory Medicine: Journal of the Forum of the European Societies of Clinical Chemistry, vol. 54, No. 7, doi:10.1515/CCLM-2015-0622, ISSN 1437-4331, (Jul. 1, 2016), pp. 1147-1155.
Agbor-Enoh, et al., “Applying rigor and reproducibility standards to assay donor-derived cell-free DNA as a non-invasive method for detection of acute rejection and graft injury after heart transplantation”, J Heart Lung Transplant, 36(9):1004-1012. doi: 10.1016/j.healun.2017.05.026. Epub May 20, 2017., 17 pages.
Agbor-Enoh, et al., “Cell-Free DNA to Detect Heart Allograft Acute Rejection”, Circulation, Mar. 23, 2021; 143(12): doi: 10.1161/CIRCULATIONAHA.120.049098. Epub Jan. 13, 2021, 1184-1197.
Ahmed, et al., “Cell Free DNA and Procalcitonin as Early Markers of Complications in ICU Patients with Multiple Trauma and Major Surgery”, Clin Lab, Dec. 1, 2016; 62(12) ; doi: 10.7754/Clin.Lab.2016.160615., 2395-2404.
Alachkar, “Serum and urinary biomarkers in acute kidney transplant rejection”, Nephrol Ther., Feb. 2012;8(1): doi: 10.1016/j.nephro.2011.07.409. Epub Oct. 21, 2011, 13-19.
Almeida, et al., “Evaluation of 16 SNPs allele-specific to quantify post hSCT chimerism by SYBR green-based qRT-PCR”, J Clin Pathol., Mar. 2013;66(3): doi: 10.1136/jclinpath-2012-201224. Epub Jan. 2, 2013., 238-242.
Andargie, et al., “Cell-free DNA maps COVID-19 tissue injury and risk of death and can cause tissue injury”, JCI Insight, Apr. 8, 2021;6(7):e147610. doi: 10.1172/jci.insight. 147610, 20 pages.
Arshad, et al., “Elevated Cell-Free Mitochondrial DNA in Filtered Plasma Is Associated with HIV Infection and Inflammation”, J Acquir Immune Defic Syndr., May 1, 2018;78(1): doi: 10.1097/QAI.0000000000001650., 111-118.
Avanzini, Stefano et al., “A mathematical model of ctDNA shedding predicts tumor detection size”, Science Advances, vol. 6, Issue eabc4308, Dec. 11, 2020, 9 pages.
Avriel, et al., “Admission Cell Free DNA Levels Predict 28-Day Mortality in Patients with Severe Sepsis in Intensive Care”, PLoS One., Jun. 23, 2014;9(6):e100514. doi: 10.1371/journal.pone.0100514. eCollection 2014., 7 pages.
Ayyadevara, et al., “Discrimination of primer 3′-nucleotide mismatch by taq DNA polymerase during polymerase chain reaction”, Anal Biochem. Aug. 15, 2000, 284(1), 11-18.
Bai, et al., “Detection and quantification of heteroplasmic mutant mitochondrial DNA by real-time amplification refractory mutation system quantitative PCR analysis: a single-step approach”, Clin Chem., Jun. 2004; 50(6); Epub Apr. 8, 2004., 996-1001.
Benn, Peter et al., “Current Controversies in Prenatal Diagnosis 2: NIPT results suggesting maternal cancer should always be disclosed”, Prenetal Diagnosis, vol. 39, No. 5, 2018, 339-343.
Bergallo, et al., “A novel TaqMAMA assay for allelic discrimination of TLR9 rs352140 polymorphism”, J Virol Methods, May 2017;243. doi: 10.1016/j.jviromet.2017.01.015. Epub Jan. 28, 2017., 25-30.
Bergallo, et al., “Evaluation of IFN-y polymorphism+874 T/A in patients with recurrent tonsillitis by PCR real time mismatch amplification mutation assay (MAMA real time PCR)”, Cytokine., Feb. 2015; 71(2): Epub Dec. 2014., 278-282.
Bewersdorf, Jan Philipp et al., “From clonal hematopoiesis to myeloid leukemia and what happens in between: Will improved understanding lead to new therapeutic and preventive opportunities?”, Blood Reviews, vol. 37, 2019, 6.
Bezieau, et al., “High incidence of N and K-Ras activating mutations in multiple myeloma and primary plasma cell leukemia at diagnosis”, Hum Mutat., Sep. 2001;18(3):. doi: 10.1002/humu.1177, 212-224.
Bianchi, Diana W. et al., “Noninvasive Prenatal Testing and Incidental Detection of Occult Maternal Malignancies”, Jama the Journal of the American Medical Association, vol. 314, No. 2, 2015, 162.
Bienkowski, et al., “Liquid biopsy for minimally invasive heart transplant monitoring: a pilot study”, J Clin Pathol., Aug. 2020; 73(8): doi: 10.1136/jclinpath-2019-205926. Epub Dec. 5, 2019., 507-510.
Blomquist, et al., “Targeted RNA-Sequencing with Competitive Multiplex-PCR Amplicon Libraries”, Plos One, 2013, vol. 8, Issue 11.
Board, et al., “Detection of PIK3CA mutations in circulating free DNA in patients with breast cancer”, Breast Cancer Res Treat, Apr. 2010;120(2): doi: 10.1007/s10549-010-0747-9. Epub Jan. 28, 2010, 461-467.
Board, et al., “Multiplexed assays for detection of mutations in PIK3CA”, Clin Chem., Apr. 2008; 54(4), 757-760.
Braun, et al., “Limitation of Circulating cfDNA Under the Use of a Cytokine Elimination Adsorber (CytoSorb) in Cardiac Surgery”, The Thoracic and Cardiovascular Surgeon, Jan. 2018; 66(S01): S1-S110, 1 page.
Bronkhorst, et al., “The emerging role of cell-free DNA as a molecular marker for cancer management”, Biomol Detect Quantif, Mar. 18, 2019;17:100087. doi: 10.1016/j.bdq.2019.100087., 23 pages.
Burgstaller, et al., “Mitochondrial DNA heteroplasmy in ovine fetuses and sheep cloned by somatic cell nuclear transfer”, BMC Dev Biol., Dec. 21, 2007; 7:141, 10 pages.
Cabel, et al., “Circulating tumor DNA changes for early monitoring of anti-PD1 immunotherapy: a proof-of-concept study”, Ann Oncol., Aug. 1, 2017; 28(8); doi: 10.1093/annonc/mdx212., 1996-2001.
Cagliani, et al., “Deoxyribonuclease Reduces Tissue Injury and Improves Survival After Hemorrhagic Shock”, J Surg Res., May 2020;249: doi: 10.1016/j.jss.2019.11.036. Epub Jan. 8, 2020., 104-113.
Castells, et al., “K-ras mutations in DNA extracted from the plasma of patients with pancreatic carcinoma: diagnostic utility and prognostic significance”, J Clin Oncol., Feb. 1999;17(2): doi: 10.1200/JCO.1999.17.2.578., 578-584.
Castleberry, et al., “Quantification of Circulating Cell-Free DNA in Pediatric Heart Transplant Recipients”, Journal of Heart and Lung Transplantation, Apr. 1, 2011; 30(4): ISSN: 1053-2498, DOI: 10.1016/j.healun.2011.01.415, S139.
Chan, et al., “Bioinformatics analysis of circulating cell-free DNA sequencing data”, Clin Biochem., Oct. 2015;48(15); doi: 10.1016/j.clinbiochem.2015.04.022. Epub May 9, 2015., 962-975.
Chang, et al., “Identification of individual DNA molecule of Mycobacterium tuberculosis by nested PCR-RLFP and capillary electrophoresis”, National Library of Medicine, 2008, 182-8.
Chen, et al., “Non-invasive prenatal diagnosis using fetal DNA in maternal plasma: a preliminary study for identification of paternally-inherited alleles using single nucleotide polymorphisms”, BMJ Open, Jul. 22, 2015;5(7):e007648. doi: 10.1136/bmjopen-2015-007648., 8 pages.
Chen, KE et al., “Multiplex PCR with the Blunt Hairpin Primers for Next Generation Sequencing”, Biotechnology and Bioprocess Engineering, vol. 22, 2017, 347-351.
Chen, Kevin et al., “Commercial ctDNA Assays for Minimal Residual Disease Detection of Solid Tumors”, Molecular Diagnosis & Therapy, vol. 25, Issue 6, Nov. 1, 2021, 757-774.
Cheng, et al., “Cell-Free DNA in Blood Reveals Significant Cell, Tissue and Organ Specific injury and Predicts COVID-19 Severity”, medRxiv., Jul. 29, 2020;2020.07.27.20163188. doi: 10.1101/2020.07.27.20163188., 16 pages.
Chiu, et al., “Effects of blood-processing protocols on fetal and total DNA quantification in maternal plasma”, Clin Chem., Sep. 2001;47(9): PubMed PMID: 11514393., 1607-1613.
Chiu, et al., “Noninvasive prenatal exclusion of congenital adrenal hyperplasia by maternal plasma analysis: a feasibility study”, Clin Chem., May 2002;48(5), 778-780.
Chu, et al., “A novel approach toward the challenge of accurately quantifying fetal DNA in maternal plasma”, Prenat Diagn., Dec. 2010;30(12-13): doi: 10.1002/pd.2656, 1226-1229.
Chung, et al., “Cell-free DNA fetal fraction and pregnancy outcome”, American Journal of Obstetrics & Gynecology, vol. 222, No. 1, 2019, S157.
Clementi, et al., “The Role of Cell-Free Plasma DNA in Critically Ill Patients with Sepsis”, Blood Purif., 2016;41(1-3): doi: 10.1159/000440975. Epub Oct. 20, 2015, 34-40.
Coombs, Catherine et al., “Therapy-Related Clonal Hematopoiesis in Patients with Non-hematologic Cancers Is Common and Associated with Adverse Clinical Outcomes”, Cell Stem Cell, vol. 21, No. 3, 2017, 374.
Daly, “Circulating donor-derived cell-free DNA: a true biomarker for cardiac allograft rejection?”, Ann Transl Med., Mar. 2015;3(4):47. doi:10.3978/j.issn.2305-5839.2015.01.35, 6 pages.
Dandel, et al., “Non-invasive cardiac allograft rejection surveillance: reliability and clinical value for prevention of heart failure”, Heart Fail Rev., Mar. 2021;26(2): doi: 10.1007/s10741-020-10023-3. Epub Sep. 5, 2020., 319-336.
Dastsooz, et al., “Multiplex ARMS PCR to Detect 8 Common Mutations of ATP7B Gene in Patients With Wilson Disease”, Hepat Mon., May 16, 2013;13(5):e8375. doi: 10.5812/hepatmon.8375. eCollection 2013., 7 pages.
De Vlaminck, et al., “Circulating cell-free DNA enables noninvasive diagnosis of heart transplant rejection”, Sci Transl Med., Jun. 18, 2014;6(241):241ra77. doi: 10.1126/scitranslmed.3007803, 20 pages.
De Vlaminck, et al., “Circulating cell-free DNA enables noninvasive diagnosis of heart transplant rejection”, Sci Transl Med., Jun. 18, 2014;6(241):241ra77. Supplemental Materials., 6 pages.
De Vlaminck, et al., “Noninvasive monitoring of infection and rejection after lung transplantation”, Proc Natl Acad Sci U S A, Oct. 27, 2015;112(43): doi: 10.1073/pnas.1517494112. Epub Oct. 12, 2015., 13336-13341.
Delgado, et al., “Characterization of cell-free circulating DNA in plasma in patients with prostate cancer”, Tumour Biol., Apr. 2013;34(2): doi: 10.1007/s13277-012-0634-6. Epub Dec. 27, 2012, 983-986.
Deshpande, et al., “Relationship Between Donor Fraction Cell-Free DNA and Treatment for Rejection in Heart Transplantation”, Pediatric Transplantation, Jun. 2022; 26(4):e14264. https://doi.org/10.1111/petr.14264, 11 pages.
Dey, et al., “A plasma telomeric cell-free DNA level in unaffected women with BRCA1 or/and BRCA2 mutations: a pilot study. Oncotarget”, Oncotarget, Dec. 29, 2017;9(3): doi: 10.18632/oncotarget.23767. eCollection Jan. 9, 2018., 4214-4222.
Dharajiya, Nilesh et al., “Incidental Detection of Maternal Neoplasia in Noninvasive Prenatal Testing”, Clinical Chemistry, vol. 64, No. 2, 2018, 329-335.
Ding, et al., “New Progress in Plasma Cell-free DNA in Clinical Applications”, Progress in Modern Biomedicine, 2016; 18: 3476, 3593-3596.
Dwivedi, et al., “Prognostic utility and characterization of cell-free DNA in patients with severe sepsis”, Crit Care, Aug. 13, 2012;16(4):R151. doi: 10.1186/cc11466., 11 pages.
Fire, et al., “Rolling replication of short DNA circles”, PNAS, 1995, 4641-4645.
Fleischhacker, et al., “Circulating nucleic acids (CNAs) and cancer—a survey”, Biochim Biophys Acta, Jan. 2007; 1775(1): doi: 10.1016/j.bbcan.2006.10.001. Epub Oct. 7, 2006., 181-232.
García Moreira, et al., “Cell-free DNA as a noninvasive acute rejection marker in renal transplantation”, Clin Chem., Nov. 2009;55(11): doi:10.1373/clinchem.2009.129072. Epub Sep. 3, 2009, 1958-1966.
Garnacho-Montero, et al., “Prognostic and diagnostic value of eosinopenia, C-reactive protein, procalcitonin, and circulating cell-free DNA in critically ill patients admitted with suspicion of sepsis”, Crit Care, Jun. 5, 2014;18(3):R116. doi: 10.1186/cc13908, 9 pages.
Ge, et al., “Haplotype block: a new type of forensic DNA markers”, Int J Legal Med, 2010, 353-361.
Ghanta, et al., “Non-invasive prenatal detection of trisomy 21using tandem single nucleotide polymorphisms”, PLoS One, Oct. 8, 2010;5(10):e13184. doi: 10.1371/journal.pone.0013184, 10 pages.
Gielis, et al., “Cell-Free DNA: An Upcoming Biomarker in Transplantation”, Am J Transplant, Oct. 2015;15(10): doi: 10.1111/ajt.13387. Epub Jul. 16, 2015, 2541-2551.
Gielis, et al., “Plasma donor-derived cell-free DNA kinetics after kidney transplantation using a single tube multiplex PCR assay”, PLoS One, 2018; 13(12): e0208207, 16 pages.
Giulio, Genovese et al., “Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence”, The New England Journal of Medicine, vol. 371, No. 26, 2014, 2477-2487.
Glaab, et al., “A novel assay for allelic discrimination that combines the fluorogenic 5′ nuclease polymerase chain reaction (TaqMan) and mismatch amplification mutation assay”, Mutat Res., Nov. 29, 1999;430(1), 1-12.
Gordon, et al., “An Algorithm Measuring Donor Cell-Free DNA in Plasma of Cellular and Solid Organ Transplant Recipients That Does Not Require Donor or Recipient Genotyping”, Front Cardiovasc Med., Sep. 22, 2016;3:33. eCollection 2016., 10 pages.
Gordon, Paul et al., “An Algorithm Measuring Donor Cell-Free DNA in Plasma of Cellular and Solid Organ Transplant Recipients That Does Not Require Donor or Recipient Genotyping”, Frontiers in Cardiovascular Medicine, 2016, vol. 3.
Gormally, et al., “Amount of DNA in plasma and cancer risk: a prospective study”, Int J Cancer, Sep. 20, 2004;111(5): doi: 10.1002/ijc.20327, 746-749.
Gotoh, et al., “Prediction of MYCN amplification in neuroblastoma using serum DNA and real-time quantitative polymerase chain reaction”, J Clin Oncol., Aug. 1, 2005;23(22): PubMed PMID: 16051962., 5205-5210.
Gripp, et al., “Homo sapiens KRAS proto-oncogene, GTPase (KRAS), RefSeqGene (LRG_344) on chromosome 12”, GenBank Submission; Accession No. NG_007524, version NG_007524.2, Aug. 16, 2020., 16 Pages.
Grskovic, et al., “Validation of a Clinical-Grade Assay to Measure Donor-Derived Cell-Free DNA in Solid Organ Transplant Recipients”, J Mol Diagn., Nov. 2016;18(6): doi: 10.1016/j.jmoldx.2016.07.003. Epub 2016, 890-902.
Guedj, et al., “A refined molecular taxonomy of breast cancer”, Oncogene, Mar. 1, 2012;31(9):1196-206. doi: 10.1038/onc.2011.301. Epub Jul. 25, 2011., 34 pages.
Hasi, et al., “Acetaldehyde dehydrogenase 2 SNP rs671 and susceptibility to essential hypertension in Mongolians: a case control study”, Genet Mol Res., Mar. 29, 2011;10(1). doi: 10.4238/vol10-1gmr1056., 537-543.
Hidestrand, et al., “Highly sensitive noninvasive cardiac transplant rejection monitoring using targeted quantification of donor-specific cell-free deoxyribonucleic acid”, J Am Coll Cardiol., Apr. 1, 2014;63(12). doi:10.1016/j.jacc.2013.09.029. Epub Oct. 16, 2013., 1224-1226.
Hidestrand, et al., “Highly Sensitive Transplant Rejection Surveillance Using Targeted Detection of Donor Specific Cell Free DNA”, J Heart Lung Transplant, Apr. 2012; 31(4), S91-S92.
Hidestrand, et al., “Influence of temperature during transportation on cellfree DNA analysis”, Fetal Diagn Ther., 2012; 31, 122-128.
Hidestrand, et al., “Quantification of Circulating Donor Specific Cell Free DNA Is an Exquisitely Sensitive Non-Invasive Indicator of Injury to the Donor Heart”, J Heart Lung Transplant, 2013; 32, S101-S102.
Hoerning, et al., “Quantitative real-time ARMS-qPCR for mitochondrial DNA enables accurate detection of microchimerism in renal transplant recipients”, Pediatr Transplant, Dec. 2011;15(8). doi: 10.1111/j.1399-3046.2011.01581.x. Epub Oct. 4, 2011, 809-818.
Hou, et al., “Application of tetra primer ARMS-PCR approach for detection of Fusarium graminearum genotypes with resistance to carbendazim”, Australian Plant Pathology, Jan. 1, 2013; 42(1), 73-78.
Huang, et al., “Circulating cell-free DNA levels correlate with postresuscitation survival rates in out-of-hospital cardiac arrest patients”, Resuscitation, Feb. 2012;83(2): doi: 10.1016/j.resuscitation.2011.07.039. Epub Aug. 22, 2011., 213-218.
Huang, et al., “Homo sapiens TSC complex subunit 1 (TSC1), RefSeqGene (LRG_486) on chromosome 9”, GenBank Submission; Accession No. NG_012386, version NG_012386.1, Sep. 21, 2020, 20 Pages.
Hudecova, “Digital PCR analysis of circulating nucleic acids”, Clin Biochem., Oct. 2015; 48(15): doi: 10.1016/j.clinbiochem.2015.03.015. Epub Mar. 28, 2015, 948-956.
Hugon, et al., “Influence of intention to adhere, beliefs and satisfaction about medicines on adherence in solid organ transplant recipients”, Transplantation., Jul. 27, 2014; 98(2): doi: 10.1097/TP.0000000000000221, 222-228.
Jaiswal, Siddhartha et al., “Clonal hematopoiesis in human aging and disease”, Science, vol. 366, No. 6465, 2019, 4.
Ji, Xing et al., “Copy number variation profile in noninvasive prenatal testing (NIPT) can identify co-existing maternal malignancies: Case reports and a literature review”, Taiwanese Journal of Obstetrics and Gynecology, vol. 57, No. 6, 2018, 871-877.
Jing, et al., “Cell-free DNA: characteristics, detection and its applications in myocardial infarction”, Curr Pharm Des., 2013;19(28): doi: 10.2174/1381612811319280012., 5135-5145.
Jordan, et al., “Donor-derived Cell-free DNA Identifies Antibody-mediated Rejection in Donor Specific Antibody Positive Kidney Transplant Recipients”, Transplant Direct, 2018; 4(9):e379, 5 pages.
Jung, et al., “Cell-free DNA in the blood as a solid tumor biomarker—a critical appraisal of the literature”, Clin Chim Acta., Nov. 11, 2010; 411(21-22): doi: 10.1016/j.cca.2010.07.032. Epub Aug. 2, 2010., 1611-1624.
Jung, Klaus et al., “Increased cell-free DNA in plasma of patients with metastatic spread in prostate cancer”, Cancer Letters, 2004, 173-180.
Kane, et al., “Application of less primer method to PCR”, DNA Polymorphism, 2004, vol. 13, pp. 34-37.
Karapetis, et al., “K-ras mutations and benefit from cetuximab in advanced colorectal cancer”, N Engl J Med., Oct. 23, 2008; 359(17). doi: 10.1056/NEJMoa0804385., 1757-1765.
Khater & Khauli, “Pseudorejection and true rejection after kidney transplantation: classification and clinical significance”, Urol Int., 90(4), 2012, 373-80.
Khush, et al., “Circulating cell-free DNA as a non-invasive marker of pediatric heart transplant rejection and immunosuppressive treatment”, J Heart Lung Transplantation, Apr. 2016. 35(4): Abstract 181,S75.
Khush, et al., “Noninvasive detection of graft injury after heart transplant using donor∧derived cell∧free DNA: A prospective multicenter study”, Am J Transplant, Oct. 2019; 19(10): doi: 10.1111/ajt.15339. Epub Apr. 8, 2019.,2889-2899.
Kim, et al., “Personalized therapy on the horizon for squamous cell carcinoma of the lung”, Lung Cancer, vol. 80, 2013, 249-255.
Kindel, et al., “Early Changes in Donor Fraction Cell-free DNA in Newly Transplanted Heart Transplant Patients”, Ishlt DF cfDNA declanation poster, 2018, 1 Page.
Kirkizlar, et al., “Detection of Clonal and Subclonal Copy-Number Variants in Cell-Free DNA from Patients with Breast Cancer Using a Massively Multiplexed PCR Methodology”, Transl Oncol., Oct. 2015; 8(5): doi: 10.1016/j.tranon.2015.08.004., 407-416.
Kirsch-Gerweck, et al., “HaploBlocks: Efficient Detection of Positive Selection in Large Population Genomic Datasets”, Mol. Biol.Evol., 2023.
Kiyomi, Morita et al., “Clearance of Somatic Mutations at Remission and the Risk of Relapse in Acute Myeloid Leukemia”, J Clin Oncol, vol. 36, No. 18, 2018, 1788-1797.
Koeppe, et al., “HIV-1-Specific CD4+ T-Cell Responses Are Not Associated With Significant Viral Epitope Variation in Persons With Persistent Plasma Viremia”, J Acquir Immune Defic Syndr, 2006, 41:140-148.
Krishnakumar, S. et al., “A comprehensive assay for targeted multiplex amplification of human DNA sequences”, PNAS, vol. 105, No. 27, Jul. 8, 2008, 9296-9301.
Kuo, et al., “Preimplantation and prenatal genetic diagnosis of aromatic L-amino acid decarboxylase deficiency with an amplification refractory mutation system-quantitative polymerase chain reaction”, Taiwan J Obstet Gynecol, Dec. 2011;50(4): doi: 10.1016/j.tjog.2011.10.012., 468-473.
Kustanovich, et al., “Life and death of circulating cell-free DNA”, Cancer Biol Ther., 2019; 20(8): doi: 10.1080/15384047.2019.1598759. Epub Apr. 16, 2019, 1057-1067.
Lajin, et al., “A quadruplex tetra-primer ARMS-PCR method for the simultaneous detection of TP53 Arg72Pro, IVS3 16bp Del/Ins and IVS6+62A>G, and NQO1 C609T polymorphisms”, Gene., Aug. 10, 2012; 504(2): Epub May 23, 2012., 268-273.
Lang, et al., “Optimized allele-specific real-time PCR assays for the detection of common mutations in KRAS and BRAF”, J Mol Diagn., Jan. 2011;13(1): doi: 10.1016/j.jmoldx.2010.11.007. Epub Dec. 23, 2010., 23-28.
Laurent-Puig, et al., “Clinical relevance of KRAS-mutated subclones detected with picodroplet digital PCR in advanced colorectal cancer treated with anti-EGFR therapy”, Clin Cancer Res., Mar. 1, 2015; 21(5): doi: 10.1158/1078-0432.CCR-14-0983. Epub Sep. 23, 2014., 1087-1097.
Lecomte, et al., “Detection of free-circulating tumor-associated DNA in plasma of colorectal cancer patients and its association with prognosis”, Int J Cancer, Aug. 10, 2002; 100(5): doi: 10.1002/ijc.10526., 542-548.
Lee, et al., “Allele-Specific Quantitative PCR for Accurate, Rapid, and Cost-Effective Genotyping”, Hum Gene Ther., Jun. 2016; 27(6): doi: 10.1089/hum.2016.011. Epub Mar. 17, 2016., 425-435.
Lefebure, et al., “Prognostic value of circulating mutant DNA in unresectable metastatic colorectal cancer”, Ann Surg., Feb. 2010;251(2): doi: 10.1097/SLA.0b013e3181c35c87, 275-280.
Lehmann-Werman, et al., “Identification of Tissue-Specific Cell Death Using Methylation Patterns of Circulating DNA”, Proc Natl Acad Sci USA, 113(13), 2016, 9 pages.
Lenaerts, Liesbeth et al., “Noninvasive Prenatal Testing and Detection of Occult Maternal Malignancies”, Clinical Chemistry, vol. 65, No. 12, 2019, 1484-1486.
Levy, et al., “Analysis of Cell-Free DNA to Assess Risk of Tumoremia Following Endoscopic Ultrasound Fine-Needle Aspiration of Pancreatic Adenocarcinomas”, Clin Gastroenterol Hepatol., Oct. 2018; 16(10): e1. doi: 10.1016/j.cgh.2018.02.048. Epub Mar. 8, 2018., 1632-1640.
Li, et al., “Multiplex co-amplification of 24 retinoblastoma gene exons after pre-amplification by long-distance PCR”, Nucleic Acids Res., Feb. 1, 1996;24(3), 538-539.
Liang, et al., “Cationic nanoparticle as an inhibitor of cell-free DNA-induced inflammation”, Nat Commun., Oct. 16, 2018;9(1):4291. doi: 10.1038/s41467-018-06603-5, 14 pages.
Lievre, et al., “KRAS mutations as an independent prognostic factor in patients with advanced colorectal cancer treated with cetuximab”, J Clin Oncol., Jan. 20, 2008; 26(3): doi: 10.1200/JCO.2007.12.5906., 374-379.
Lin, et al., “A new diagnostic system for ultra-sensitive and specific detection and quantification of Candidatus Liberibacter asiaticus, the bacterium associated with citrus Huanglongbing”, J Microbial Methods, 2010, 17-25.
Liu, et al., “ABO chimerism determined by real-time polymerase chain reaction analysis after ABO-incompatible haematopoietic stem cell transplantation”, Blood Tranfus, Jan. 2013; 11(1): doi: 10.2450/2012.0013-12. Epub Jul. 4, 2012., 43-52.
Liu, et al., “Comparison of next-generation sequencing systems”, J Biomed Biotechnol., 2012; 2012: doi: 10.1155/2012/251364. Epub Jul. 5, 2012., 1-11.
Livergood, “Adverse perinatal outcomes and cell free DNA no calls: Beyond low fetal fraction”, American Journal of Obstetrics & Gynecology, vol. 218, No. 1, 2018, S169.
Lizardi, et al., “Mutation detection and single-molecule counting using isothermal rolling-circle amplification”, Nature Genetics, 1998, 225-232.
Llop, et al., “Development of a highly sensitive nested-PCR procedure using a single closed tube for detection of Erwinia amylovora in asymptomatic plant material”, Appl Environ Microbial., 2000, 2071-8.
Lo, et al., “Plasma placental RNA allelic ratio permits noninvasive prenatal chromosomal aneuploidy detection”, Nat Med., Feb. 2007; 13(2): doi: 10.1038/nm1530. Epub Jan. 7, 2007., 218-223.
Lo, et al., “Transplantation monitoring by plasma DNA sequencing”, Clin Chem., Jul. 2011;57(7): doi: 10.1373/clinchem.2011.166686. PubMed PMID: 21566070., 941-942.
Luo, et al., “Detection of usual and atypical aldehyde dehydrogenase alleles by mismatch amplification mutation assay”, Clin Chem Lab Med., Dec. 2001;39(12): doi: 10.1515/CCLM.2001.189., 1195-1197.
Maheswaran, S. et al., “Detection of Mutations in EGFR in Circulating Lung-Cancer Cells”, N Engl J Med, vol. 359, No. 4, Jul. 24, 2008, 366-377.
Mak, et al., “Rapid diagnosis of Wilson disease by a 28-mutation panel: real-time amplification refractory mutation system in diagnosing acute Wilsonian liver failure”, Clin Chim Acta., Dec. 2008; 398(1-2): doi: 10.1016/j.cca.2008.08.002. Epub Aug. 8, 2008., 39-42.
Mamanova, L. et al., “Target-enrichment strategies for next-generation sequencing”, Nat Methods, vol. 7, No. 2, 2010, 111-118.
Mamun, et al., “The Escherichia coli UVM response is accompanied by an SOS-independent error-prone DNA replication activity demonstrable in vitro”, Molecular Microbiology, 2000, 368-380.
Manage, et al., “Genotyping single nucleotide polymorphisms in human genomic DNA with an automated and self-contained PCR cassette”, J Mol Diagn., Sep. 2014; 16(5): doi:10.1016/j.jmoldx.2014.04.004. Epub Jul. 2, 2014., 550-557.
Margulies, et al., “Genome Sequencing in Open Microfabricated High Density Picoliter Reactors”, Nature, 437(7057), 2005, 376-380.
Martinez-Herrero, et al., “Cancer protection elicited by a single nucleotide polymorphism close to the adrenomedullin gene”, J Clin Endocrinol Metab., Apr. 2013; 98(4): doi: 10.1210/jc.2012-4193. Epub Feb. 28, 2013., E807-E810.
Mehra, et al., “Gene expression profiles and B-type natriuretic peptide elevation in heart transplantation: more than a hemodynamic marker”, Circulation, Jul. 4, 2006;114(1 Suppl), I21-I26.
Mehra, et al., “International Society for Heart and Lung Transplantation working formulation of a standardized nomenclature for cardiac allograft vasculopathy-2010”, J Heart Lung Transplant, Jul. 2010; 29(7) .doi: 10.1016/j.healun.2010.05.017., 717-727.
Mengel, et al., “The molecular phenotype of heart transplant biopsies: relationship to histopathological and clinical variables”, Am J Transplant, Sep. 2010; 10(9): doi: 10.1111/j.1600-6143.2010.03182.x., 2105-2115.
Metzker, Michael, Declaration of Michael L. Metzker, Ph.D. from IPR2018-01317, 2004.
Misale, et al., “Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer”, Nature, Jun. 28, 2012; 486(7404): doi: 10.1038/nature11156., 532-536.
Mouliere, et al., “Circulating Cell-Free DNA from Colorectal Cancer Patients May Reveal High KRAS or BRAF Mutation Load”, Transl Oncol., Jun. 1, 2013; 6(3): doi: 10.1593/tlo.12445. Print Jun. 2013., 319-328.
Mueller, P. R. et al., “In Vivo Footprinting of a Muscle Specific Enhancer by Ligation Mediated PCR”, Science, vol. 249, Nov. 10, 1989, 780-786.
Myers, et al., “ACB-PCR quantification of somatic oncomutation”, Methods Mol Biol., 2014;1105: doi:10.1007/978-1-62703-739-6_27, 345-363.
Nakamura, et al., “Ex Vivo Liver Perfusion with Arterial Blood from a Pig with Ischemic Liver Failure”, Artificial Organs, 1999, 153-160.
Newton, et al., “Analysis of any point mutation in DNA. The amplification refractory mutation system (ARMS)”, Nucleic Acids Res., Apr. 11, 1989;17(7): doi: 10.1093/nar/17.7.2503, 2503-2516.
No Author Listed, “Jury Rules in Favor of Natera, Finding All Asserted Patents Valid and Infrindged by ArcherDX/Invitae; Awards $19.35 Million in Past Damages for Royalties and Lost Profits”, Natera Press Release, 2023, 4 pgs.
No Author Listed, “Natera Inc. vs. ArcherDx Verdict Form, Case 1:20-cv-00125-GBW”, 2023, 1-12.
No Author Listed, “The Journal of Heart and Lung Transplantation”, Apr. 2012., vol. 31, Issue 4, Supplement, pp. A1-A4, S1-S310. https://www.google.de/searchq=The+Journal+of+Heart+and+Lung+Transplantation+Volume+31,+Issue+4,+Supplement&sourceid=ie7&rls=com.microsoft:en-US:IE-Address&ie=&oe=#spf=1604593918239, Last Accessed: Oct. 13, 2015., A1-A4.
No Author Listed, NIH, “Quantitative Detection of Circulating Donor-Specific DNA in Organ Transplant Recipients (DTRT-Multi-Center Study) (DTRT)”, ClinicalTrials.gov Identifier: NCT02109575., Apr. 10, 2014, Last updated Mar. 26, 2021, 9 pages.
North, et al., “Cell-free DNA donor fraction analysis in pediatric and adult heart transplant patients by multiplexed allele-specific quantitative PCR: Validation of a rapid and highly sensitive clinical test for stratification of rejection probability”, PLoS One, Jan. 13, 2020; 15(1):e0227385. doi: 10.1371/journal.pone.0227385. eCollection 2020., 48 pages.
Norton, et al., “Perinatal and genetic outcomes associated with no call cfDNA results in 18,496 pregnancies”, American Journal of Obstetrics & Gynecology, vol. 224, No. 2, 2021, S3.
Nui, et al., “The Functional Integrity of a Normothermic Perfusion System Using Artificial Blood in Pig Liver”, Journal of Surgical Research, 2006, 189-198.
Oeth, et al., “Qualitative and quantitative genotyping using single base primer extension coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MassARRAY®).”, Methods Mol. Biol., 2009; 578, 307-343.
Orou, et al., “Allele-specific competitive blocker PCR: a one-step method with applicability to pool screening”, Hum Mutat., 1995; 6(2): doi: 10.1002/humu.1380060209., 163-169.
Parsons, et al., “Allele-specific competitive blocker-PCR detection of rare base substitution”, Methods Mol Biol., 2005;291, 235-245.
Partpart-Li, Sonya et al., “The Effect of Preservative and Temperature on the Analysis of Circulating Tumor DNA”, Clin Cancer Res, 2017, 2471-2477.
PCT/US2017/059808, “International Preliminary Report on Patentability”, mailed May 16, 2019, 8 pages.
PCT/US2017/059808, “International Search Report and Written Opinion for Application”, mailed Jan. 25, 2018, 12 pages.
PCT/US2018/038598, “International Preliminary Report on Patentability”, mailed Jan. 2, 2020, 6 pages.
PCT/US2018/038598, “International Search Report and Written Opinion”, mailed Sep. 7, 2018, 8 pages.
PCT/US2018/038609, “International Preliminary Report on Patentability”, mailed Jan. 2, 2020, 7 pages.
PCT/US2018/038609, “International Search Report and Written Opinion”, mailed Sep. 10, 2018, 9 pages.
PCT/US2018/065845, “International Preliminary Report on Patentability mailed Jun. 25, 2020”, WIPO, 9 pages.
PCT/US2018/065845, “International Search Report and Written Opinion mailed Mar. 8, 2019”, WIPO, 13 pages.
Peng, et al., “Comparison of K-ras mutations in lung, colorectal and gastric cancer”, Oncol Lett., Aug. 2014; 8(2): doi: 10.3892/ol.2014.2205. Epub May 30, 2014., 561-565.
Peng, Q et al., “Reducing amplification artifacts in high multiplex amplicon sequencing by using molecular barcodes”, BMC Genomics, vol. 16. No. 586, 2015, 12 pages.
Peyster, et al., “Advanced Morphologic Analysis for Diagnosing Allograft Rejection: The Case of Cardiac Transplant Rejection”, Transplantation, Aug. 2018;102(8): doi: 10.1097/TP.0000000000002189., 1230-1239.
Price, et al., “Cost-effective interrogation of single nucleotide polymorphisms using the mismatch amplification mutation assay and capillary electrophoresis”, Electrophoresis, Dec. 2010; 31(23-24): doi: 10.1002/elps.201000379., 3881-3888.
Purhonen, et al., “Human plasma cell-free DNA as a predictor of infectious complications of neutropenic fever in hematological patients”, Infect Dis (Lond)., Apr. 2015; 47(4): doi: 10.3109/00365548.2014.985711. Epub Feb. 9, 2015., 255-259.
Qin, et al., “Quantitative assessment of hematopoietic chimerism by quantitative real-time polymerase chain reaction of sequence polymorphism systems after hematopoietic stem cell transplantation”, Chin Med J (Engl), Aug. 2011; 124(15), 2301-2308.
Quail, et al., “A tale of three next generation sequencing platforms: comparison of Ion torrent, pacific biosciences and illumina MiSeq sequencers”, BMC Genomics, Jul. 24, 2012; 13:341. doi: 10.1186/1471-2164-13-341, 13 pages.
Ragalie, et al., “Description of Longitudinal Measurement of Donor Fraction of Cell-Free DNA and Correlation to Clinical Outcomes”, ISHLT poster, 2018, 1 page.
Ragalie, et al., “Noninvasive Assay for Donor Fraction of Cell-Free DNA in Pediatric Heart Transplant Recipients”, J Am Coll Cardiol., Jun. 26, 2018;71(25): doi: 10.1016/j.jacc.2018.04.026, 2982-2983.
Reinert, et al., “Analysis of Plasma Cell-Free DNA by Ultradeep Sequencing in Patients with Stages I to UI Colorectal Cancer”, JAMA Oncology, 2019, 1-74.
Richmond, et al., “Donor fraction cell-free DNA and rejection in adult and pediatric heart transplantation”, J Heart Lung Transplant, May 2020;39(5): doi: 10.1016/j.healun.2019.11.015. Epub Nov. 29, 2019., 454-463.
Roedder, et al., “Biomarkers in solid organ transplantation: establishing personalized transplantation medicine”, Genome Med., Jun. 8, 2011;3(6):37, 12 pages.
Sairafi, et al., “Donor Cell Composition and Reactivity Predict Risk of Acute Graft-versus-Host Disease After Allogeneic Hematopoietic Stem Cell Transplantation”, J Immunol Res., 2016, 11 pages.
Sanmamed, et al., “Quantitative cell-free circulating BRAFV600E mutation analysis by use of droplet digital PCR in the follow-up of patients with melanoma being treated with BRAF inhibitors”, Clin Chem., Jan. 2015; 61(1): doi: 10.1373/clinchem.2014.230235. Epub Nov. 19, 2014., 297-304.
Sapio, et al., “Detection of BRAF mutation in thyroid papillary carcinomas by mutant allele-specific PCR amplification (MASA)”, Eur J Endocrinol., Feb. 2006;154(2): doi: 10.1530/eje.1.02072, 341-348.
Saukkonen, et al., “Cell-free plasma DNA as a predictor of outcome in severe sepsis and septic shock.”, Clin Chem., Jun. 2008; 54(6): doi: 10.1373/clinchem.2007.101030. Epub Apr. 17, 2008. PubMed PMID: 18420731., 1000-1007.
Scheffer, et al., “Association between low fetal fraction in cell-free DNA testing and adverse pregnancy outcome: A systematic review”, Prenatal Diagnosis, vol. 41, No. 10, 2021, 1287-1295.
Schnittger, et al., “Development and validation of a real-time quantification assay to detect and monitor BRAFV600E mutations in hairy cell leukemia”, Blood., Mar. 29, 2012; 119(13): doi: 10.1182/blood-2011-10-383323. Epub Feb. 13, 2012., 3151-3154.
Schutz, et al., “Graft-derived cell-free DNA, a noninvasive early rejection and graft damage marker in liver transplantation: A prospective, observational, multicenter cohort study”, PLoS Med., Apr. 25, 2017; 14(4):e1002286. doi: 10.1371/journal.pmed.1002286. eCollection Apr. 2017., 19 pages.
Schwarzenbach, et al., “Cell-free nucleic acids as biomarkers in cancer patients”, Nat Rev Cancer, Jun. 2011; 11(6): doi: 10.1038/nrc3066. Epub May 12, 2011, 426-437.
Scott, et al., “Elevated nuclear and mitochondrial cell-free deoxyribonucleic acid measurements are associated with death after infant cardiac surgery”, J Thorac Cardiovasc Surg., Aug. 2022; 164(2): doi: 10.1016/j.jtcvs.2021.10.066. Epub Dec. 24, 2021., 367-375.
Scott, et al., “Total Cell-Free DNA Predicts Death and Infection Following Pediatric and Adult Heart Transplantation”, Ann Thorac Surg., Oct. 2021; 112(4): doi: 10.1016/j.athoracsur.2020.08.006. Epub Oct. 8, 2020., 1282-1289.
Sefrioui, et al., “Clinical value of chip-based digital-PCR platform for the detection of circulating DNA in metastatic colorectal cancer”, Dig Liver Dis., Oct. 2015; 47(10): doi: 10.1016/j.dld.2015.05.023. Epub Jun. 5, 2015, 884-890.
Sheffield, et al., “Attachment of a 40-base-pair G + C-rich sequence (GC-clamp) to genomic DNA fragments by the polymerase chain reaction results in improved detection of single-base changes”, Proc Natl Acad Sci U S A, Jan. 1989;86(1), 232-236.
Shi, et al., “Development of a single multiplex amplification refractory mutation system PCR for the detection of rifampin-resistant Mycobacterium tuberculosis”, Gene., Nov. 1, 2013; 530(1): Epub Aug. 19, 2013, 95-99.
Shimabukuro-Vornhagen, et al., “Cytokine release syndrome”, J Immunother Cancer, Jun. 15, 2018; 6(1):56. doi: 10.1186/s40425-018-0343-9, 14 pages.
Sigdel, et al., “A rapid noninvasive assay for the detection of renal transplant injury”, Transplantation, Jul. 15, 2013;96(1): doi: 10.1097/TP.0b013e318295ee5a., 97-101.
Sigdel, Tara et al., “Optimizing Detection of Kidney Transplant Injury by Assessment of Donor-Derived Cell-Free DNA via Massively Multiplex PCR”, Journal of Clinical Medicine, vol. 8, No. 1, 2018, 19.
Sigdel, Tara et al., “Plasma Donor-Derived Cell-Free DNA Quantification by massively multiplex PCR Distinguishes Kidney Transplant Acute Rejection”, Transplantation, vol. 102, No. 7s, 2018, s178-s179.
Singh, et al., “Aspergillus infections in transplant recipients”, Clin Microbiol Rev., Jan. 2005;18(1), 44-69.
Snyder, et al., “Universal noninvasive detection of solid organ transplant rejection”, Proc Natl Acad Sci U S A, Apr. 12, 2011;108(15); doi: 10.1073/pnas.1013924108. Epub Mar. 28, 2011. PubMed PMID: 21444804; PubMed Central PMCID: PMC3076856., 6229-6234.
Sparks, et al., “Noninvasive prenatal detection and selective analysis of cell-free DNA obtained from maternal blood: evaluation for trisomy 21 and trisomy 18”, Am J Obstet Gynecol., Apr. 2012; 206(4): doi: 10.1016/j.ajog.2012.01.030. Epub Jan. 26, 2012, 319.e1-319.e9.
Sparks, et al., “Selective analysis of cell-free DNA in maternal blood for evaluation of fetal trisomy”, Prenat Diagn., Jan. 2012; 32(1). Epub Jan. 6, 2012., 3-9.
Spindler, et al., “Cell-free DNA in healthy individuals, noncancerous disease and strong prognostic value in colorectal cancer”, Int J Cancer, Dec. 15, 2014; 135(12): doi: 10.1002/ijc.28946. Epub Jun. 17, 2014, 2984-2991.
Spindler, et al., “KRAS-mutated plasma DNA as predictor of outcome from irinotecan monotherapy in metastatic colorectal cancer”, Br J Cancer, Dec. 10, 2013; 109(12). doi: 10.1038/bjc.2013.633. Epub Nov. 21, 2013., 3067-3072.
Spindler, et al., “Quantitative cell-free DNA, KRAS, and BRAF mutations in plasma from patients with metastatic colorectal cancer during treatment with cetuximab and irinotecan”, Clin Cancer Res., Feb. 15, 2012; 18(4). doi: 10.1158/1078-0432.CCR-11-0564. Epub Jan. 6, 2012., 1177-1185.
Steensma, D. P. et al., “Clonal hematopoiesis of indeterminate potential and its distinction from myelodysplastic syndromes”, Blood, vol. 126, No. 1, 2015, 9-16.
Stein, “Next-Generation Sequencing Update”, Genetic Engineering & Biotechnology News, Sep. 1, 2008; 28(15). https://www.genengnews.com/magazine/97/next-generation-sequencing-update/, 10 pages.
Steinborn, et al., “Coexistence of Bos taurus and B. indicus mitochondrial DNAs in nuclear transfer-derived somatic cattle clones”, Genetics, Oct. 2002;162(2), 823-829.
Stemmer, et al., “Use of magnetic beads for plasma cell-free DNA extraction: toward automation of plasma DNA analysis for molecular diagnostics”, Clin Chem., Nov. 2003;49(11): PubMed PMID: 14578335., 1953-1955.
Strausberg, et al., “Homo sapiens placenta-specific 4, mRNA (cDNA clone MGC:120720 IMAGE:7939530), complete cds”, GenBank Submission; Accession No. BC093685, version BC093685.1., Jan. 18, 2007, 2 Pages.
Strohmeier, et al., “Multiplex genotyping of KRAS point mutations in tumor cell DNA by allele-specific real-time PCR on a centrifugal microfluidic disk segment”, Microchimica Acta., 2014;181 (13-14), 1681-1688.
Suzuki, et al., “Characterization of circulating DNA in healthy human plasma”, Clin Chim Acta., Jan. 2008;387(1-2): doi: 10.1016/j.cca.2007.09.001. Epub Sep. 8, 2007., 55-58.
Swinkels, et al., “Effects of blood-processing protocols on cell-free DNA quantification in plasma”, Clin Chem., Mar. 2003;49(3): PubMed PMID: 12600978, 525-526.
Tabernero, et al., “Analysis of circulating DNA and protein biomarkers to predict the clinical activity of regorafenib and assess prognosis in patients with metastatic colorectal cancer: a retrospective, exploratory analysis of the Correct trial”, Lancet Oncol., Aug. 2015; 16(8): doi: 10.1016/S1470-2045(15)00138-2. Epub Jul. 13, 2015., 937-948.
Taira, et al., “Novel high-speed droplet-allele specific-polymerase chain reaction: application in the rapid genotyping of single nucleotide polymorphisms”, Clin Chim Acta., Sep. 23, 2013; 424: doi: 10.1016/j.cca.2013.04.024. Epub May 17, 2013., 39-46.
Taira, et al., “Quantitative monitoring of single nucleotide mutations by allele-specific quantitative PCR can be used for the assessment of minimal residual disease in patients with hematological malignancies throughout their clinical course”, Clin Chim Acta., Jan. 14, 2011; 412(1-2): doi: 10.1016/j.cca.2010.09.011. Epub Sep. 16, 2010., 53-58.
Takai, et al., “Clinical utility of circulating tumor DNA for molecular assessment in pancreatic cancer”, Sci Rep., Dec. 16, 2015; 5:18425. doi: 10.1038/srep18425., 10 pages.
Taly, et al., “Multiplex picodroplet digital PCR to detect KRAS mutations in circulating DNA from the plasma of colorectal cancer patients”, Clin Chem., Dec. 2013; 59(12): doi: 10.1373/clinchem.2013.206359. Epub Aug. 12, 2013., 1722-1731.
Tamkovich, et al., “Circulating nucleic acids in blood of healthy male and female donors”, Clin Chem., Jul. 2005; 51(7): PubMed PMID: 15976134., 1317-1319.
Tanem, et al., “Abstract 16873: Association of Preoperative Cell-Free DNA Levels and Outcome Following Pediatric Cardiopulmonary Bypass”, Circulation, Nov. 17, 2020; 142(S3): https://doi.org/10.1161/circ.142.suppl_3.16873., 1-6.
Thermofisher Scientific, “How Ion AmpliSeq Targeted Sequencing Technology Works”, https://www.thermofisher.com/us/en/home/life-science/sequencing/next-generation-sequencing/ion-torrent-next-generation-sequencing-workflow/ion-torrent-next-generation-sequencing-select-targets/ampliseq-target-selection/how-ampliseq-technology-work.
Thierry, et al., “A Targeted Q-PCR-Based Method for Point Mutation Testing by Analyzing Circulating DNA for Cancer Management Care”, Methods Mol Biol., 2016;1392: doi: 10.1007/978-1-4939-3360-0_1, 1-16.
Thierry, et al., “Clinical validation of the detection of KRAS and BRAF mutations from circulating tumor DNA”, Nat Med., Apr. 2014; 20(4): doi: 10.1038/nm.3511. Epub Mar. 23, 2014., 430-435.
Tomita-Mitchell, et al., “Human gene copy number spectra analysis in congenital heart malformations”, Physiol Genomics, May 1, 2012; 44(9): doi: 10.1152/physiolgenomics.00013.2012. Epub Feb. 7, 2012., 518-541.
Tong, et al., “Diagnostic developments involving cell-free (circulating) nucleic acids”, Clin Chim Acta., Jan. 2006; 363(1-2): Epub Aug. 26, 2005. Review. PubMed PMID: 16126188, 187-196.
Van Orsouw, et al., “Rapid design of denaturing gradient-based two-dimensional electrophoretic gene mutational scanning tests”, Nucleic Acids Res., May 15, 1998; 26(10), 2398-2406.
Van Raemdonck, et al., “Ex-vivo lung perfusion”, Transplant International, vol. 28, Issue 6, 2014, 643-656.
Vandekerkhove, G et al., “Circulating Tumor DNA Reveals Clinically Actionable Somatic Genome of Metastatic Bladder Cancer”, Clinical Cancer Research, 2017, 6487-6497.
Vannucchi, et al., “A quantitative assay for JAK2(V617F) mutation in myeloproliferative disorders by ARMS-PCR and capillary electrophoresis”, Leukemia, Jun. 2006; 20(6), 1055-1060.
Verhoeven, et al., “Biomarkers to Assess Graft Quality During Conventional and Machine Preservation in Liver Transplantation”, J Hepatol., 2014, 672-84.
Veseloskva, “The use of cell-free nucleic acids in maternal plasma for non-invasive prenatal diagnosis of monogenic diseases, placental insufficiency-related complications and Down syndrome”, Thesis from Charles University in Prague, 2011, 104 pages.
Wang, et al., “DNA Degradation Test Predicts Success in Whole-Genome Amplification from Diverse Clinical Samples”, Journal of Molecular Diagnostics, vol. 9, 2007, 441-451.
Wangkumhang, et al., “WASP: a Web-based Allele-Specific PCR assay designing tool for detecting SNPs and mutations”, BMC Genomics, Aug. 14, 2007;8:275, 9 Pages.
Wapner, et al., “Expanding the scope of noninvasive prenatal testing: detection of fetal microdeletion syndromes”, Am J Obstet Gynecol., Mar. 2015; 212(3): doi: 10.1016/j.ajog.2014.11.041. Epub Dec. 2, 2014., 332.e1-332.e9.
Wilkins, et al., “IMP PCR primers detect single nucleotide polymorphisms for Anopheles gambiae species identification, Mopti and Savanna rDNA types, and resistance to dieldrin in Anopheles arabiensis”, Malar J., Dec. 19, 2006; 5:125., 7 pages.
Wood, et al., “Molecular histology of lung cancer: From targets to treatments”, Cancer Treatment Reviews, vol. 41, 2015, 361-375.
Woude, et al., “Methods of identifying drugs with selective effects against cancer cells”, Oct. 7, 1997, Nucleic acid sequence search reports AC: 151794, Accession I51796., 2 Pages.
Xie, et al., “Designing highly multiplex PCR primer sets with Simulated Annealing Design using Dimer Likelihood Estimation (SADDLE)”, Nat Commun., vol. 13, No. 1, 2022, 1881.
Yamada, et al., “Detection of K-ras gene mutations in plasma DNA of patients with pancreatic adenocarcinoma: correlation with clinicopathological features”, Clin Cancer Res., Jun. 1998; 4(6), 1527-1532.
Yamauchi Medical Clinic, “Chromosome abnormality”, http://www.yamauchi-iin.com/kaisetu/1241.htm, (Dec. 10, 2015 updated), Dec. 10, 2015, 3 pages.
Yi, et al., “PCR/LDR/capillary electrophoresis for detection of single-nucleotide differences between fetal and maternal DNA in maternal plasma”, Prenat Diagn., Mar. 2009; 29(3): doi: 10.1002/pd.2072., 217-222.
Zangwill, et al., “Effect of endomyocardial biopsy on levels of donor-specific cell-free DNA”, J Heart Lung Transplant, Oct. 2019; 38(10): doi: 10.1016/j.healun.2019.06.005. Epub Jun. 28, 2019., 1118-1120.
Zhang, et al., “A novel multiplex tetra-primer ARMS-PCR for the simultaneous genotyping of six single nucleotide polymorphisms associated with female cancers”, PLoS One, Apr. 17, 2013; 8(4):e62126. doi: 10.1371/journal.pone.0062126. Print 2013., 8 pages.
Zheng, et al., “Whole-exome sequencing to identify novel somatic mutations in squamous cell lung cancers”, International Journal of Oncology, vol. 43, 2015, 755-764.
Goessl, C. et al., “DNA Alterations in Body Fluids as Molecular Tumor Markers for Urological Malignancies”, European Urology, vol. 41, 2002, 668-676.
Kaper, Fiona et al., “Abstract 1164: Parallel preparation of targeted resequencing libraries from 480 genomic regions using multiplex PCR on the Access Array system”, American Association for Cancer Research, 2010, 1-2.
Kaper, Fiona et al., “Parallel Preparation of Targeted Resequencing Libraries from 480 Genomic Regions Using Multiplex PCR on the Access Array System”, Fluidigm Poster, 2011, 1.
Vargas, D. Y. et al., “Multiplex Real-Time PCR Assays that Measure the Abundance of Extremely Rare Mutations Associated with Cancer”, PLOS One, vol. 11, No. 5, May 31, 2016, 26 pgs.
Wong, I. H. et al., “Quantitative Analysis of Tumor-derived Methylated p161NK4a Sequences in Plasma, Serum, and Blood Cells of Hepatocellular Carcinoma Patients”, Clinical Cancer Research, vol. 9, Mar. 2003, 1047-1052.
European Application No. 014198110, European Search Report Mailed Apr. 28, 2015, 3 pages.
PRNewswire (Research Suggests Daily Consumption of Orange Juice Can Reduce Blood Pressure and May Provide Beneficial Effects to Blood Vessel Function: New Study Identified Health Benefits in Orange Juice, Dec. 8, 2010), 3 pages.
“Abstracts for CNAPS III Circulating Nucleic Acids in Plasma and Serum and Serum Proteomics”, Clinical Chemistry, vol. 49, No. 11, 2003, 33 pages.
“Abstracts for CNAPS IV Circulating Nucleic Acids in Plasma/Serum”, Fourth International Conference on Circulating Nucleic Acids in Plasma/Serum (CNAPS-IV), 2005, 40 pages.
“How Many Carbs in a Potato?, [Online]”, New Health Guide, Nov. 1, 2014, 3 pages.
“Random variable”, The Penguin Dictionary of Mathematics, 2008, 1 page.
“Fixed Medium”, “Academic Press”, http://www.xreferplus.com/entry.do?pp=1&id=310, 1996, 1 pg.
Abaan, O. D. et al., “The Exomes of the NCI-60 Panel: A Genomic Resource for Cancer Biology and Systems Pharmacology”, Cancer Res., vol. 73, No. 14, Jul. 15, 2013, 4372-4382.
Abbosh, C. et al., “Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution”, Nature, vol. 545, May 25, 2017, 446-451.
Abd-Elsalam, Kamel A. , “Bioinformatic Tools and Guideline for PCR Primer Design”, African Journal of Biotechnology, vol. 2, 2003, pp. 91-95.
Abidi, S. et al., “Leveraging XML-based electronic medical records to extract experiential clinical knowledge: An automated approach to generate cases for medical case-based reasoning systems”, International Journal of Medical Informatics, 68(1-3), 2002, 187-203.
Adalsteinsson, V. A. et al., “Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors”, Nature Communications, vol. 18, No. 1324, 2017, 13 pages.
Adinolfi, M. et al., “Rapid Detection of Aneuploidies by Microsatellite and the Quantitative Fluorescent Polymerase Chain Reaction”, Prenatal Diagnosis, vol. 17, No. 13, 1997, 1299-1311.
Agarwal, Ashwin. et al., “Commercial Landscape of Noninvasive Prenatal Testing in the United States”, Prenatal Diagnosis,33, 2013, 521-531.
Agbor-Enoh, S. et al., “Donor-derived cell-free DNA predicts allograft failure and mortality after lung transplantation”, EBioMedicine, vol. 40, 2019, 541-553.
Alaeddini, R. et al., “Forensic implications of genetic analyses from degraded DNA—A review”, Forensic Science International: Genetics, vol. 4, 2010, 148-157.
Alberts, B. et al., “Chapter 20: Germ Cells and Fertilization”, Molecular Biology of the Cell, Fourth Edition, 2002, 1127-1156.
Alberts, B. et al., “Chapter 4: DNA and Chromosomes”, Molecular Biology of the Cell, Fourth Edition, 2002, 191-234.
Alizadeh, Mehdi et al., “Quantitative Assessment of Hematopoietic Chimerism after Bone Marrow Transplantation by Real-time Quantitative Polymerase Chain Reaction”, Blood, vol. 99, No. 12, Jun. 15, 2002, 4618-4625.
Alkan, Can et al., “Personalized Copy Number and Segmental Duplication Maps Using Next-Generation Sequencing”, Nature Genetics, 41, 10, 2009, 1061-1068.
Allaire, F R., “Mate selection by selection index theory”, Theoretical Applied Genetics, 57(6), 1980, 267-272.
Allan, J. et al., “Micrococcal Nuclease Does Not Substantially Bias Nucleosome Mapping”, Journal of Molecular Biology, vol. 417, Jan. 30, 2012, 152-164.
Allawi, Hatim T. et al., “Thermodynamics of internal C⋅T Mismatches in DNA”, Nucleic Acids Research, 26 (11), 1998, 2694-2701.
Ambardar, S. et al., “High Throughput Sequencing: An Overview of Sequencing Chemistry”, Indian J. Microbiol., vol. 56, No. 4, 2016, 394-404.
Amicucci, P. et al., “Prenatal Diagnosis of Myotonic Dystrophy Using Fetal DNA Obtained from Maternal Plasma”, Clinical Chemistry, vol. 46, No. 2, 2000, 301-302.
Andras, S. C. et al., “Strategies for Signal Amplification in Nucleic Acid Detection”, Molecular Biotechnology, vol. 19, 2001, 29-44.
Anker, P. et al., “Circulating DNA in Plasma or Serum”, Medicina, vol. 60, 2000, 699-702.
Anker, P. et al., “Detection of circulating tumour DNA in the blood (plasma/serum) of cancer patients”, Cancer and Metastasis Reviews, vol. 18, 1999, 65-73.
Anker, P. et al., “The Second International Symposium on Circulating Nucleic Acids in Plasma and Serum (CNAPS-2) held in conjunction with the 6th Annual Scientific Symposium of the Hong Kong Cancer Institute”, Clinical Chemistry, vol. 47, No. 2, 2001, 361-370.
Ansorge, Wilhelm J., “Next-generation DNA Sequencing Techniques”, New Biotechnology, vol. 25, No. 4, Feb. 2, 2009, 195-203.
Antonarakis, S. E. et al., “Chromosome 21 and Down Syndrome: From Genomics to Pathophysiology”, Nature Reviews Genetics, vol. 5, Oct. 2004, 725-738.
Aoki, Yasuhiro, “Statistical and Probabilistic Bases of Forensic DNA Testing”, The Journal of the Iwate Medical Association, 2002, vol. 54, p. 81-94.
Arandjelovic, M. et al., “Two-Step Multiplex Polymerase Chain Reaction improves the Speed and Accuracy of Genotyping Using DNA from Noninvasive and Museum Samples”, Molecular Ecology Resources, vol. 9, 2009, pp. 28-36.
Ashoor, G. et al., “Fetal fraction in maternal plasma cell-free DNA at 11-13 weeks' gestation: relation to maternal and fetal characteristics”, Ultrasound in Obstetrics and Gynecology, vol. 41, 2013, 26-32.
Ashoor, Ghalia et al., “Chromosome-Selective Sequencing of Maternal Plasma Cell-Free DNA for First-Trimester Detection of Trisomy 21 and Trisomy 18”, American Journal of Obstetrics & Gynecology, 206, 2012, 322.e1-322.e5.
Ashoor, Ghalia et al., “Fetal Fraction in Maternal Plasma Cell-Free DNA at 11-13 Weeks' Gestation: Effect of Maternal and Fetal Factors”, Fetal Diagnosis Therapy, 2012, 1-7.
Auld, D. S., “Use of Chelating Agents to Inhibit Enzymes”, Methods in Enzymology, vol. 158, 1988, 110-114.
Avent, Neil D. et al., “Cell-free Fetal DNA in The Maternal Serum and Plasma: Current and Evolving Applications”, Current Opinion in Obstretrics and Gynecology, vol. 21, No. 2, Apr. 1, 2009, 175-179.
Avgidou, K. et al., “Prospective first-trimester screening for trisomy 21 in 30,564 pregnancies”, American Journal of Obstetrics and Gynecology, vol. 192, 2005, 1761-1767.
Ayala, et al., “Long-Term Follow-Up of Donor Chimerism Tolerance After Human Liver Transplantation”, Liver Transplantation, vol. 15, No. 6,, May 28, 2009, 581-591.
Bada, Michael A. et al., “Computational Modeling of Structural Experimental Data”, Methods in Enzymology,317, 2000, 470-491.
Bai, H. et al., “Detection and Clinical Significance of Intratumoral EGFR Mutational Heterogeneity in Chinese Patients with Advanced Non-Small Cell Lung Cancer”, PLOS One, vol. 8, No. 2, Feb. 2013, 7 pages.
Balavoine, Guillaume, “Identification of Members of Several Homeobox Genes in a Planarian Using a Ligation-Mediated Polymerase Chain Reaction Technique”, Nucleic Acids Research, vol. 24, 1996, pp. 1547-1553.
Balduini, et al., “Utility of Biochemical Markers in the Follow-up Heart Transplant Recipients”, Transplantation Proceedings, vol. 35, No. 8, Dec. 1, 2003, 3075-3078.
Bale, J. R. et al., “Reducing Birth Defects: Meeting the Challenge in the Developing World”, Institute of Medicine of the National Academies, 2003, 270 pgs.
Ballif, B. C. et al., “Detection of Low-Level Mosaicism by Array CGH in Routine Diagnostic Specimens”, American Journal of Medical Genetics Part A, vol. 140A, 2006, 2757-2767.
Banfi, G. et al., “The role of ethylenediamine tetraacetic acid (EDTA) as in vitro anticoagulant for diagnostic purposes”, Clin. Chem., vol. 45, No. 5, 2007, 565-576.
Barbazuk, et al., “SNP Discovery via 454 Transcriptome Sequencing”, The Plant Journal, vol. 51, Jul. 27, 2007, 910-918.
Barra, G. B. et al., “EDTA-mediated inhibition of DNases protects circulating cell-free DNA from ex vivo degradation in blood samples”, Clinical Biochemistry, vol. 48, 2015, 976-981.
Barski, A. et al., “High-Resolution Profiling of Histone Methylations in the Human Genome”, Cell, vol. 129, May 18, 2007, 823-837.
Bartlett, John M. et al., “PCR Protocols”, PCR Protocols, vol. 226, 2003, 519 pages.
Bashashati, A. et al., “Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling”, Journal of Pathology, vol. 231, 2013, 21-34.
Bau, Stephan et al., “Targeted next-generation sequencing by specific capture of multiple genomic loci using low-volume microfluidic DNA arrays”, Anal Bioanal Chem, vol. 393, 2009, 171-175.
Bauer, M. et al., “A prospective analysis of cell-free fetal DNA concentration in maternal plasma as an indicator for adverse pregnancy outcome”, Prenatal Diagnosis, vol. 26, 2006, 831-836.
Baxter, L. L. et al., “Discovery and genetic localization of Down syndrome cerebellar phenotypes using the Ts65Dn mouse”, Human Molecular Genetics, vol. 9, No. 2, Jan. 2000, 195-202.
Baxter-Lowe, et al., “Tracking Microchimeric DNA in Plasma to Diagnose and Manage Organ Transplant Rejection”, Clinical Chemistry, vol. 52, No. 4, Apr. 1, 2006, 559-561.
Beaumont, Mark A et al., “The Bayesian Revolution in Genetics”, Nature Reviews Genetics, 5, 2004, 251-261.
Beck, et al., “Next Generation Sequencing of Serum Circulating Nucleic Acids from Patients with Invasive Ductal Breast Cancer Reveals Differences to Healthy and Nonmalignant Controls”, Molecular Cancer Research, vol. 8, No. 3, Mar. 1, 2010, 335-342.
Beck, J. et al., “Digital Droplet PCR for Rapid Quantification of Donor DNA in the Circulation of Transplant Recipients as a Potential Universal Biomarker of Graft Injury”, Clinical Chemistry, vol. 59, No. 12, 2013, 1732-1741.
Beck, J. et al., “Profile of the Circulating DNA in Apparently Healthy Individuals”, Clinical Chemistry, vol. 55, No. 4, 2009, 730-738.
Beer, Alan E. et al., “The Biological Basis of Passage of Fetal Cellular Material into the Maternal Circulation”, Annals New York Academy of Sciences, 731, 1994, 21-35.
Beerenwinkel, et al., “Methods for Optimizing Antiviral Combination Therapies”, Bioinformatics, 19(1), 2003, i16-i25.
Beerenwinkel, N. et al., “Geno2pheno: estimating phenotypic drug resistance from HIV-1 genotypes”, Nucleic Acids Research, 31(13), 2003, 3850-3855.
Belostotsky, Dmitry A. et al., “Plant Systems Biology”, Methods in Molecular Biology, vol. 553, Aug. 25, 2009, 3-408.
Bender, et al., “A Multiplex SNP Typing Approach for the DNA Pyrosequencing Technology”, International Congress Series, vol. 1288, Apr. 20, 2006, 73-75.
Benjamini, Y. et al., “Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing”, Journal of the Royal Statistical Society, Series B (Methodological), vol. 57, No. 1, 1995, 289-300.
Benn, P. et al., “Non-Invasive Prenatal Testing for Aneuploidy: Current Status and Future Prospects”, Ultrasound Obstet Gynecol, 42, 2013, 15-33.
Benn, P et al., “Non-Invasive prenatal Diagnosis for Down Syndrome: the Paradigm Will Shift, but Slowly”, Ultrasound Obstet. Gynecol., 39, 2012, 127-130.
Bennett, S. T. et al., “Toward the $1000 human genome”, Pharmacogenomics, vol. 6, No. 4, 2005, 373-382.
Bentley, et al., “High-resolution, High-throughput HLA Genotyping by Next-generation Sequencing”, Tissue Antigens, vol. 74, No. 5, Nov. 1, 2009, 393-403.
Bentley, David R et al., “Accurate Whole Human Genome Sequencing Using Reversible Terminator Chemistry”, Nature, 456, 6, 2008, 53-59.
Bergen, A. W. et al., “Effects of DNA mass on multiple displacement whole genome amplification and genotyping performance”, BMC Biotechnology, vol. 5, No. 24, Sep. 16, 2005, 11 pgs.
Bermudez, M. et al., “Single-cell sequencing and mini-sequencing for preimplantation genetic diagnosis”, Prenatal Diagnosis, 23, 2003, 669-677.
Beroud, C. et al., “Prenatal diagnosis of spinal muscular atrophy by genetic analysis of circulating fetal cells”, The Lancet, vol. 361, Mar. 22, 2003, 1013-1014.
Bevinetto, Gina, Bevinetto (5 Foods All Pregnant Women Need, American Baby, available at http://www.parents.com/pregnancy/mybody/nutrition/5greatpregnancyfoods/, Apr. 15, 2008), 8 pgs.
Bianchi, D W. et al., “Fetal gender and aneuploidy detection using fetal cells maternal blood: analysis of NIFTY I data”, Prenat Diagn 2002; 22, 2002, 609-615.
Bianchi, D W. et al., “Insights Into Fetal and Neonatal Development Through Analysis of Cell-Free RNA in Body Fluids”, Early Human Development, vol. 86, No. 11, Nov. 2010, 747-752.
Bianchi, D. W., “Circulating Fetal DNA: Its Origin and Diagnostic Potential—A Review”, Placenta, vol. 25, Supplemental A, May 2004, S93-S101.
Bianchi, D. W. et al., “Genome-Wide Fetal Aneuploidy Detection by Maternal Plasma DNA Sequencing”, Obstetrics & Gynecology, vol. 119, No. 5, May 2012, 890-901.
Bianchi, D. W., “Review: Fetal Cells in the Maternal Circulation: Feasibility for Prenatal Diagnosis”, British Journal of Haematology, vol. 105, 1999, 574-583.
Binladen, J. et al., “The Use of Coded PCR Primers Enables High-Throughput Sequencing of Multiple Homolog Amplification Products by 454 Parallel Sequencing”, PLOS One, Issue 2, Feb. 2007, 9 pages.
Birch, Lyndsey et al., “Accurate and Robust Quantification of Circulating Fetal and Total DNA in Maternal Plasma from 5 to 41 Weeks of Gestation”, Clinical Chemistry, 51(2), 2005, 312-320.
Birkenkamp-Demtroder, K. et al., “Abstract 3653: Sequencing of plasma cfDNA from patients with locally advanced bladder cancer for surveillance and therapeutic efficacy monitoring”, Cancer Research, vol. 78, No. 13 Supplement, Jul. 2019, 1 page.
Bischoff, F. Z. et al., “Cell-free fetal DNA in maternal blood: kinetics, source and structure”, Human Reproduction Update, vol. 11, No. 1, 2005, 59-67.
Bischoff, F. Z. et al., “Intact fetal cells in maternal plasma: are they really there?”, Lancet, vol. 361, 2003, 139-140.
Bisignano, et al., “PGD and Aneuploidy Screening for 24 Chromosomes: Advantages and Disadvantages of Competing Platforms”, Reproductive BioMedicine Online, 23, 2011, 677-685.
Blomquist, T M. et al., “Targeted RNA-Sequencing with Competitive Multiplex—PCR Amplicon Libraries”, Plos One, vol. 8, Issue 11, Nov. 2013, 14 pages.
Blow, N., “The personal side of genomics”, Nature, vol. 449, Oct. 4, 2007, 627-630.
Board, R.E. et al., “Detection of BRAF mutations in the tumour and serum of patients enrolled in the AZD6244 (ARRY-142886) advanced melanoma phase II study”, British Journal of Cancer, vol. 101, 2009, 1724-1730.
Bodenreider, O., “The Unified Medical Language System (UMLS): Integrating Biomedical Terminology”, Nucleic Acids Research, 32, (Database issue), 2004, D267-D270.
Bordoni, et al., “Evaluation of Human Gene Variant Detection in Amplicon Pools by the GS-FLX Parallel Pyrosequencer”, BMC Genomics, vol. 9, Oct. 8, 2008, 1-8.
Boudsocq, F. et al., “Sulfolobus solfataricus P2 DNA polymerase IV (Dpo4): an archael DinB-like DNA polymerase with lesion-bypass properties akin to eukaryotic poln”, Nucleic Acids Research, vol. 29, No. 22, 2001, 4607-4616.
Bouma, B. N. et al., “Human Blood Coagulation Factor”, The Journal of Biological Chemistry, vol. 252, No. 18, 1977, 6432-6437.
Brastianos, P. K. et al., “Genomic Characterization of Brain Metastases Reveals Branched Evolution and Potential Therapeutic Targets”, Cancer Discovery, vol. 5, Sep. 26, 2015, 1164-1177.
Breithaupt, Holger, “The Future of Medicine”, EMBO Reports, 21(61), 2001, 465-467.
Brinza, D. et al., “2SNP: scalable phasing based on 2-SNP haplotypes”, Bioinformatics, vol. 22, No. 3, 2006, 371-373.
Brockman, et al., “Quality Scores and SNP Detection in Sequencing-by-synthesis Systems”, Genome Research, vol. 18, No. 5, May 1, 2008, 763-770.
Broude, N E. et al., “High-Level Multiplex DNA Amplification”, Antisense & Nucleic Acid Drug Development, vol. 11, 2001, 327-332.
Broude, N. E. et al., “High Multiplexity PCR Based on PCR Suppression”, DNA Amplification Current Technologies and Applications, 2004, 61-76.
Broude, N. E. et al., “Multiplex Allele-specific Target Amplification based on PCR Suppression”, PNAS, vol. 98, No. 1, Jan. 2, 2001, 206-211.
Brownie, Jannine et al., “The Elimination of Primer-Dimer Accumulation in PCR”, Nucleic Acids Research, 25(16), 1997, 3235-3241.
Browning, S. R. et al., “Rapid and Accurate Haplotype Phasing and Missing-Data Inference for Whole-Genome Association Studies by Use of Localized Haplotype Clustering”, The American Journal of Human Genetics, vol. 81, Nov. 2007, 1084-1097.
Bryant, A. P., “Terminology of Sugars”, Ind. Eng. Chem., vol. 26, No. 2, 1933, 231.
Burkey, B. F. et al., “Hepatic apolipoprotein J is secreted as a lipoprotein”, Journal of Lipid Research, vol. 33, 1992, 1517-1526.
Burkova, E. E. et al., “Extremely Stable Soluble High Molecular Mass Multi-Protein Complex with DNase Activity in Human Placental Tissue”, PLOS One, vol. 9, No. 11: e011234, Nov. 26, 2014, 26 pages.
Burnham, P. et al., “Myriad Applications of Circulating Cell-Free DNA in Precision Organ Transplant Monitoring”, Annals of the American Thoracic Society, vol. 14, Supplement 3, Sep. 2017, S237-S241.
Bustamante-Aragones, Ana et al., “New Strategy for The Prenatal Detection/Exclusion of Paternal Cystic Fibrosis Mutations in Maternal Plasma”, Journal of Cystic Fibrosis, vol. 7, Issue 6, Nov. 1, 2008, 505-510.
Butler, et al., “Cardiovascular Magnetic Resonance in the Diagnosis of Acute Heart Transplant Rejection: A Review”, Journal of Cardiovascular Magnetic Resonance, vol. 11, No. 1, Mar. 12, 2009, 1-11.
Butler, J. et al., “The Development of Reduced Size STR Amplicons as Tools for Analysis of Degraded DNA*”, Journal of Forensic Sciences, vol. 48, No. 5, 2003, 1054-1064.
Butt, A. N. et al., “Overview of Circulating Nucleic Acids in Plasma/Serum: Update on Potential Prognostic and Diagnostic Value in Diseases Excluding Fetal Medicine and Oncology”, Ann. N.Y. Acad. Sci., vol. 1137, 2008, 236-242.
Cairns, Paul et al., “Homozygous Deletions of 9p21 in Primary Human Bladder Tumors Detected by Comparative Multiplex Polymerase Chain Reaction”, Cancer Research, 54, 1994, 1422-1424.
Caliendo, Angela, “Multiplex PCR and Emerging Technologies for the Detection of Respiratory Pathogens”, Clinical Infectious Diseases, 52(4), 2011, S326-S330.
Calin, G. A. et al., “A MicroRNA Signature Associated with Prognosis and Progression in Chronic Lymphocytic Leukemia”, N Engl J Med, vol. 353, 2005, 1793-1801.
Campbell, P. J. et al., “Subclonal phylogenetic structures in cancer revealed by ultra-deep sequencing”, PNAS, vol. 105, No. 35, Sep. 2, 2008, 13081-13086.
Canick, J. A. et al., “The impact of maternal plasma DNA fetal fraction on next generation sequencing tests for common fetal aneuploidies”, Prenatal Diagnosis, vol. 33, 2013, 667-674.
Cansar, “Hs-578-T—Copy Number Variation—Cell Line Synopsis”, ICR Cancer Research UK, Retrieved on Mar. 26, 2018 from https://cansar.icr.ac.uk/cansar/cell-lines/Hs-578-T/copy_number_variation/chromosome_8/, Mar. 26, 2018, 50 pgs.
Cao, Y. et al., “Clinical Evaluation of Branched DNA Signal Amplification for Quantifying HIV Type 1 in Human Plasma”, AIDS Research and Human Retroviruses, vol. 11, No. 3, 1995, 353-361.
Carnevale, Alessandra et al., “Attitudes of Mexican Geneticists Towards Prenatal Diagnosis and Selective Abortion”, American Journal of Medical Genetics, 75, 1998, 426-431.
Carvalho, B. et al., “Exploration, normalization, and genotype calls of high-density oligonucleotide SNP array data”, Biostatistics, vol. 8, No. 2, 2007, 485-499.
Casbon, J. A. et al., “A method for counting PCR template molecules with application to next-generation sequencing”, Nucleic Acids Research, vol. 39, No. 12, Apr. 13, 2011, 1-8.
Castleberry, C. D. et al., “Quantification of Circulating Cell—Free DNA in Pediatric Heart Transplant Recipients”, Journal of Heart and Lung Transplantation, vol. 30, No. 4, Apr. 1, 2011, S139.
Chakraborty, R. et al., “Paternity Exclusion by DNA Markers: Effects of Paternal Mutations”, Journal of Forensic Sciences, vol. 41, No. 4, Jul. 1996, 671-677.
Chan, Allen K. et al., “Cell-free Nucleic Acids in Plasma, Serum and Urine: A New Tool in Molecular Diagnosis”, Annals of Clinical Biochemistry, vol. 40, Issue 2, Mar. 1, 2003, 122-130.
Chan, K.C. et al., “Size Distributions of Maternal and Fetal DNA in Maternal Plasma”, Clinical Chemistry, vol. 50, No. 1, 2004, 88-92.
Chang, H.W. et al., “Assessment of Plasma DNA Levels, Allelic Imbalance, and CA 125 as Diagnostic Tests for Cancer”, Journal of the National Cancer Institute, vol. 94, No. 22, Nov. 20, 2002, 1697-1703.
Chavali, Sreenivas et al., “Oligonucleotide Properties Determination And Primer Designing: A Critical Examination of Predictions”, Bioinformatics, vol. 21, 2005, pp. 3918-3925.
Chen, E. et al., “Noninvasive Prenatal Diagnosis of Fetal Trisomy 18 and Trisomy 13 by Maternal Plasma DNA Sequencing”, PLoS ONE, 6 (7), e21791, 2011, 7 pgs.
Chen, Bing-Yuan et al., “PCR Cloning Protocols”, PCR Cloning Protocols, vol. 192, 2002, 434 pages.
Chen, C. P. et al., “Fetal DNA in maternal plasma: the prenatal detection of a paternally inherited fetal aneuploidy”, Prenatal Diagnosis, vol. 20, 2000, 353-357.
Chen, X. Q. et al., “Microsatallite alterations in plasma DNA of small cell lung cancer patients”, Nature Medicine, vol. 2, No. 9, Sep. 1996, 1033-1035.
Chetty, Shilpa et al., “Uptake of Noninvasive Prenatal Testing (NIPT) in Women Following Positive Aneuploidy Screening”, Prenatal Diagnosis,33, 2013, 542-546.
Cheung, S. W. et al., “Rapid Publication: Microarray-Based CGH Detects Chromosomal Mosaicism Not Revealed by Conventional Cytogenetics”, American Journal of Medical Genetics Part A, vol. 143A, 2007, 1679-1686.
Cheung, V. G. et al., “Whole genome amplification using a degenerate oligonucleotide primer allows hundreds of genotypes to be performed on less than one nanogram of genomic DNA”, Proceedings of the National Academy of Sciences, USA, vol. 93, Dec. 1996, 14676-14679.
Chim, S. S. et al., “Detection and Characterization of Placental MicroRNAs in Maternal Plasma”, Clinical Chemistry, vol. 54, No. 3, 2008, 482-490.
Chinnapapagari, S. K. et al., “Treatment of Maternal Blood Samples with Formaldehyde Does Not Alter the Proportion of Circulatory Fetal Nucleic Acids (DNA and mRNA) in Maternal Plasma”, Clinical Chemistry, vol. 51, No. 3, 2005, 653-655.
Chitty, L. S. et al., “Noninvasive Prenatal Screening for Genetic Diseases Using Massively Parallel Sequencing of Maternal Plasma DNA”, Cold Spring Harbor Perspectives in Medicine, vol. 5, No. 9, 2015, 20 pages.
Chiu, R. et al., “Non-Invasive Prenatal Assessment of Trisomy 21 by Multiplexed Maternal Plasma DNA Sequencing: Large Scale Validity Study”, BMJ, 342, c7401, 2011, 9 pgs.
Chiu, R.W.K. et al., “Hypermethylation of RASSF1A in Human and Rhesus Placentas”, The American Journal of Pathology, vol. 170, No. 3, Mar. 2007, 941-950.
Chiu, Rossa W. et al., “Effects of Blood-Processing Protocols on Fetal and Total DNA Quantification in Maternal Plasma”, Clinical Chemistry, 47(9), 2001, 1607-1613.
Chiu, Rossa W.K. et al., “Maternal Plasma DNA Analysis with Massively Parallel Sequencing by Litigation for Noninvasive Prenatal Diagnosis of Trisomy 21”, Clinical Chemistry, 56, 3, 2010, 459-463.
Chiu, Rossa W.K. et al., “Non-Invasive Prenatal Diagnosis by Single Molecule Counting Technologies”, Trends in Genetics, 25 (7), 2009, 324-331.
Chiu, Rossa W.K. et al., “Noninvasive Prenatal Diagnosis of Fetal Chromosomal Aneuploidy by Massively Parallel Genomic Sequencing of DNA in Maternal Plasma (with Supporting Information)”, PNAS, vol. 105, No. 51, 2008, 20458-20463.
Choi, M. et al., “Genetic diagnosis by whole exome capture and massively parallel DNA sequencing”, PNAS, vol. 106, No. 45, Nov. 10, 2009, 19096-19101.
Choi, Y. et al., “Comparison of phasing strategies for whole human genomes”, PLOS Genetics, Apr. 5, 2018, 26 pages.
Chu, T. et al., “Statistical Considerations for Digital Approaches to Non-Invasive Fetal Genotyping”, Bioinformatics (Advance Access publication), 26 (22), 2010, 2863-2866.
Chu, Tianjiao et al., “Statistical Model for Whole Genome Sequencing and its Application to Minimally Invasive Diagnosis of Fetal Genetic Disease”, Bioinformatics, 25(10), 2009, 1244-1250.
Chu, Tianjiao et al., “A Novel Approach Toward the Challenge of Accurately Quantifying Fetal DNA in Maternal Plasma”, Prenatal Diagnosis,30, 2010, 1226-1229.
Chung, G. T. et al., “Lack of Dramatic Enrichment of Fetal DNA in Maternal Plasma by Formaldehyde Treatment”, Clinical Chemistry, vol. 51, No. 3, 2005, 655-658.
Church, et al., “Multiplex DNA Sequencing”, Science, vol. 240, No. 4849, Apr. 8, 1988, 185-188.
Ciriello, G. et al., “Emerging landscape of oncogenic signatures across human cancers”, Nature Genetics, vol. 45, No. 10, Oct. 2013, 1127-1135.
Clausen, F. B. et al., “Improvement in fetal DNA extraction from maternal plasma. Evaluation of the NucliSens Magnetic Extraction system and the QIAamp DSP Virus Kit in comparison with the QIAamp DNA Blood Mini Kit”, Prenatal Diagnosis, vol. 27, 2007, 6-10.
Cole, Neal W. et al., “Hyperglycemia-Induced Membrane Lipid Peroxidation and Elevated Homocysteine Levels Are Poorly Attenuated by Exogenous Folate in Embryonic Chick Brains”, Comparative Biochemistry and Physiology, Part B, 150, 2008, 338-343.
Colella, S. et al., “QuantiSNP: an Objectives Bayes Hidden-Markov Model to Detect and Accurately Map Copy Number Variation Using SNP Genotyping Data”, Nucleic Acids Research, 35 (6), 2007, 2013-2025.
Conlin, L. K. et al., “Mechanisms of mosaicism, chimerism and uniparental disomy identified by single nucleotide polymorphism array analysis”, Human Molecular Genetics, vol. 19, No. 7, Jan. 6, 2010, 1263-1275.
Coombes, R. C., “Abstract P4-01-02: Early detection of residual breast cancer through a robust, scalable and personalized analysis of circulating tumour DNA (ctDNA) antedates overt metastatic recurrence”, Cancer Research, vol. 79, No. 4 Supplement, Feb. 15, 2019.
Cossu, Gianfranco et al., “Rh D/d Genotyping by Quantitative Polymerase Chain Reaction and Capillary Zone Electrophoresis”, Electrophoresis, 17, 1996, 1911-1915.
Couraud, S. et al., “Noninvasive Diagnosis of Actionable Mutations by Deep Sequencing of Circulating Free DNA in lung Cancer from Never-Smokers: A Proof-of-Concept Study from BioCAST / IFCT-1002”, Clinical Cancer Research, vol. 20, No. 17, Jul. 10, 2014, 4613-4624.
Couraud, S. et al., “Supplementary Data for Noninvasive Diagnosis of Actionable Mutations by Deep Sequencing of Circulating Free DNA in lung Cancer from Never-Smokers: A Proof-of-Concept Study from BioCAST / IFCT-1002”, 2014, 13 pages.
Coyle, J. F. et al., “Standards for detailed clinical models as the basis for medical data exchange and decision support”, International Journal of Medical Informatics, 69(2-3), 2003, 157-174.
Craig, D. W. et al., “Identification of genetic variants using bar-coded multiplexed sequencing”, Nature Methods, vol. 5, Oct. 2008, 887-893.
Crespo-Leiro, et al., “Gene Expression Profiling for Monitoring Graft Rejection in Heart Transplant Recipients”, Transplantation Proceedings, vol. 41, No. 6, Jul. 1, 2009, 2240-2243.
Cronn, R. et al., “Multiplex sequencing of plant chloroplast genomes using Solexa sequencing-by-synthesis technology”, Nucleic Acids Research, vol. 36, No. 19, Aug. 27, 2008, 11 pgs.
Cross, Jillian et al., “Resolution of trisomic mosaicism in prenatal diagnosis: estimated performance of a 50K SNP microarray”, Prenat Diagn 2007; 27, 2007, 1197-1204.
Cunningham, K. S. et al., “An approach to endomyocardial biopsy interpretation”, Journal of Clinical Pathology, vol. 59, No. 2, Mar. 2006, 121-129.
Dahl, et al., “Multigene Amplification and Massively Parallel Sequencing for Cancer Mutation Discovery”, Proceedings of the National Academy of Sciences, vol. 104, No. 22, May 29, 2007, 9387-9392.
Dambrin, et al., “A New Rejection Criteria in the Heterotopically Placed Rat Heart by Non-invasive Measurement of Dp/Dtmax”, The Journal of Heart and Lung Transplantation, vol. 18, No. 6, Jun. 18, 1999, 524-531.
D'Aquila, Richard et al., “Maximizing sensitivity and specificity of PCR by pre-amplification heating”, Nucleic Acids Research, 19(13), 1991, p. 3749.
Daruwala, Raoul-Sam et al., “A Versatile Statistical Analysis Algorithm to Detect Genome Copy Number Variation”, PNAS, 101(46), 2004, 16292-16297.
Dawson, S.J. et al., “Analysis of Circulating Tumor DNA to Monitor Metastatic Breast Cancer”, The New England Journal of Medicine, vol. 368, No. 13, Mar. 28, 2013, 1199-1209.
De Bruin, E. et al., “Spatial and temporal diversity in genomic instability processes defines lung cancer evolution”, Science, vol. 346, No. 6206, Oct. 10, 2014, 251-256.
De Jong, M. M. et al., “Genes other than BRCA 1 and BRCA2 involved in breast cancer susceptibility”, J. Med. Genet., vol. 39, 2009, 225-242.
De Vries, et al., “Diagnostic genome profiling in mental retardation”, Am J Hum Genet, 77, published online Aug. 30, 2005, 2005, 606-616.
Deangelis, M. et al., “Solid-phase Reversible Immobilization for the Isolation of PCR Products”, Nucleic Acids Research, 23 (22), 1995, 4742-4743.
Deb, Mahua et al., “Development of a Multiplexed PCR Detection Method for Barley and Cereal Yellow Dwarf Viruses, Wheat Spindle Streak Virus, Wheat Streak Mosaic Virus and Soil-Borne Wheat Mosaic Virus”, Journal of Virological Methods, vol. 148, 2008, pp. 17-24.
Delaneau, O. et al., “Shape-IT: new rapid and accurate algorithm for haplotype inference”, BMC Bioinformatics, vol. 9, No. 540, Dec. 16, 2008, 14 pages.
Delgado, P. O. et al., “Characterization of cell-free circulating DNA in plasma in patients with prostate cancer”, Tumor Biol., vol. 34, 983-986, 2013.
Deng, S. et al., “TNER: A Novel Background Error Suppression Method for Mutation Detection in Circulating Tumor DNA”, bioRxiv, http://dx.doi.org/10.1101/214379, Nov. 5, 2017, 12 pgs.
Deutsch, S. et al., “Detection of aneuploidies by paralogous sequence quantification”, J Med Genet, vol. 41, 2004, 908-915.
Devaney, S. et al., “Noninvasive Fetal Sex Determination Using Cell-Free Fetal DNA: A Systematic Review and Meta-analysis”, JAMA, 306 (6), 2011, 627-636.
Dhallan, et al., “Methods to Increase the Percentage of Free Fetal DNA Recovered from the Maternal Circulation”, JAMA, 291(9), 2004, 1114-1119.
Dhallan, Ravinder et al., “A non-invasive test for prenatal diagnosis based on fetal DNA present in maternal blood: a preliminary study”, The Lancet, 369, 2007, 474-481.
Di, X. et al., “Dynamic model based algorithms for screening and genotyping”, Bioinformatics, vol. 21, No. 9, 2005, 1958-1963.
Dias-Santagata, D. et al., “BRAF V600E Mutations Are Common in Pleomorphic Xanthoastrocytoma: Diagnostic and Therapeutic Implications”, PLoS One, vol. 6, No. 3, Mar. 2011, 9 pages.
Dickover, R. E. et al., “Optimization of Specimen-Handling Procedures for Accurate Quantitation of Levels of Human Immunodeficiency Virus RNA in Plasma by Reverse Transcriptase PCR”, Journal of Clinical Microbiology, vol. 36, No. 4, 1998, 1070-1073.
Dieffenbach, C W. et al., “General concepts for PCR primer design”, Genome Research. PCR methods and Applications vol. 3, 1993, S30-S37.
Diehl, F. et al., “Circulating mutant DNA to assess tumor dynamics”, Nature Medicine, vol. 14, No. 9, Jul. 31, 2008, 985-990.
Diehl, F. et al., “Detection and quantification of mutations in the plasma of patients with colorectal tumors”, PNAS, vol. 102, No. 45, Nov. 8, 2005, 16368-16373.
Dietmaier, W. et al., “Multiple Mutation Analyses in Single Tumor Cells with Improved Whole Genome Amplification”, American Journal of Pathology, vol. 154, No. 1, Jan. 1999, 83-95.
Ding, C et al., “Direct molecular haplotyping of long-range genomic DNA with M1-PCR”, PNAS 100(13), 2003, 7449-7453.
Ding, C. et al., “MS analysis of single-nucleotide differences in circulating nucleic acids: Application to noninvasive prenatal diagnosis”, PNAS, vol. 101, No. 29, Jul. 20, 2004, 10762-10767.
Dodge, Y., “Bayes' Theorem”, The Concise Encyclopedia of Statistics, 2008, 30-31.
Dohm, J. et al., “Substantial Biases in Ultra-Short Read Data Sets From High-Throughput DNA Sequencing”, Nucleic Acids Research, 36 (16), e105, 2008, 10 pgs.
Dolganov, Gregory et al., “A Novel Method of Gene Transcript Profiling in Airway Biopsy Homogenates Reveals Increased Expression of a Na-K+ -Cl-Cotransporter (NKCC1) in Asthmatic Subjects”, Genome Res., 11, 2001, 1473-1483.
Donaghue, C. et al., “Detection of mosaicism for primary trisomies in prenatal samples by QF-PCR and karyotype analysis”, Prenatal Diagnosis, vol. 25, 2005, 65-72.
Donohoe, Gerard G et al., “Rapid Single-Tube Screening of the C282Y Hemochromatosis Mutation by Real-Time Multiplex Allele-specific PCR without Fluorescent Probes”, Clinical Chemistry, 46, 10, 2000, 1540-1547.
Donoso, P. et al., “Current Value of Preimplantation Genetic Aneuploidy Screening in IVF”, Human Reproduction Update, 13(1), 2007, 15-25.
Doostzadeh, et al., “High Throughput Automated Allele Frequency Estimation by Pyrosequencing”, PLoS ONE, vol. 3, No. 7, Jul. 16, 2008, 1-4.
Dorit, D. L., “cDNA Amplification Using One-sided (Anchored) Pcr”, Current Protocols in Molecular Biology, vol. 17, 1992, pp. 15.6.1-15.6.10.
Dorit, Robert L. et al., “One-sided Anchored Polymerase Chain Reaction for Amplification and Sequencing of Complementary DNA”, Methods in Enzymology, vol. 218 1993, pp. 36-47.
Dowd, P. et al., “On the mechanism of the anticlotting action of vitamin R quinone”, Proc. Natl. Acad. Sci. USA, vol. 92, 1995, 8171-8175.
Downward, J., “Targeting Ras Signalling Pathways in Cancer Therapy”, Nature Reviews, vol. 3, Jan. 2003, 11-22.
Dressman, D. et al., “Transforming single DNA molecules into fluorescent magnetic particles for detection and enumeration of genetic variations”, PNAS, vol. 100, No. 15, Jul. 22, 2003, 8817-8822.
Echeverri, et al., “Caffeine's Vascular Mechanisms of Action”, International Journal of Vascular Medicine vol. 2010(2010), 10 pages, Aug. 25, 2010.
Edwards, M. C. et al., “Multiplex PCR: Advantages, Development, and Applications”, Genome Research, vol. 3, 1994, S65-S75.
Efron, B. et al., “Bootstrap Methods for Standard Errors, Confidence Intervals, and Other Measures of Statistical Accuracy”, Statistical Science, vol. 1, No. 1, 1986, 54-77.
Ehrich, Mathias et al., “Noninvasive Detection of Fetal Trisomy 21 by Sequencing of DNA in Maternal Blood: A Study in a Clinical Setting”, American Journal of Obstetrics & Gynecology, 204, 2011, 205.e1-205.e11.
Eichler, H, “Mild Course of Fetal Rh D Haemolytic Disease due to Maternal Alloimmunisation to Paternal HLA Class I and II Antigens”, Vox Sang, 68, 1995, 243-247.
Ellison, Aaron M., “Bayesian Inference in Ecology”, Ecology Letters, vol. 7, 2004, 509-520.
Ellonen, P. et al., “Development of SNP Microarray for Supplementary Paternity Testing”, International Congress Series, 1261, 2004, 12-14.
Elnifro, Elfath M., “Multiplex PCR: Optimization and Application in Diagnostic Virology”, Clinical Microbiology Reviews, vol. 13, 2000, pp. 559-570.
Eltoukhy, H. et al., “Modeling and Base-Calling for DNA Sequencing-By-Synthesis”, IEEE, 2006, II-1032-II-1035.
EP06838311.6, “European Communication and Extended European Search Report”, mailed Dec. 30, 2008, 8 pgs.
EP08742125.1, “European Communication pursuant to Article 94(3) EPC and Examination Report”, mailed Feb. 12, 2010, 5 pgs.
Erijman, Ariel et al., “Transfer-PCR (TPCR): A Highway for DNA Cloning and Protein Engineering”, Journal of Structural Biology, vol. 175, 2011, pp. 171-177.
Erlich, R. L. et al., “Next-generation sequencing for HLA typing of class loci”, BMC Genomics, vol. 12, No. 42, 2011, 13 pages.
Eronen, L. et al., “HaploRec: efficient and accurate large-scale reconstruction of haplotypes”, BMC Bioinformatics, vol. 7, No. 542, Dec. 22, 2006, 18 pages.
European Commission, “The 7th International Conference on Circulating Nucleic Acids in Plasma and Serum (CNAPS VII) in Madrid—Spain”, The International Conference on Circulating Nucleic Acids in Plasma and Serum, Oct. 24, 2011, 2 pgs.
Everitt, B. S., “Medical Statistics From A to Z”, 2003, 3 pages.
Fackenthal, J. D. et al., “Aberrant RNA splicing and its functional consequences in cancer cells”, Disease Models & Mechanisms, vol. 1, 2008, 37-42.
Faham, M. et al., “Deep-sequencing approach for minimal residual disease detection in acute lymphoblastic leukemia”, Blood Journal, vol. 120, No. 26, Dec. 20, 2012, 5173-5180.
Falcon, O., “Screening for trisomy 21 by fetal tricuspid regurgitation, nuchal translucency and maternal serum free b-hCG and PAPP-A at 11 + 0 to 13 + 6 weeks”, Ultrasound Obstet Gynecol, vol. 27, 2006, 151-155.
Fan, et al., “Whole-genome molecular haplotyping of single cells”, Nature Biotechnology, vol. 29, No. 1, Jan. 1, 2011, 51-57.
Fan, C H. et al., “Detection of Aneuploidy with Digital Polymerase Chain Reaction”, Analytical Chemistry, vol. 79, No. 19, Oct. 1, 2007, 7576-7579.
Fan, Christina H. et al., “Non-Invasive Prenatal Measurement of the Fetal Genome”, Nature, doi:10.1038/nature11251, 2012, 26 pgs.
Fan, Christina H et al., “Noninvasive Diagnosis of Fetal Aneuploidy by Shotgun Sequencing DNA from Maternal Blood”, PNAS, 105, 42, 2008, 16266-16271.
Fan, H. C. et al., “In Principle Method for Noninvasive Determination of the Fetal Genome”, Nat. Prec., 2010, 16 pgs.
Fan, H. C. et al., “Microfluidic digital PCR enables rapid prenatal diagnosis of fetal aneuploidy”, American Journal of Obstetrics & Gynecology, vol. 200, May 2009, 543.e1-543.e7.
Fan, H. Christina et al., “Sensitivity of Noninvasive Prenatal Detection of Fetal Aneuploidy from Maternal Plasma Using Shotgun Sequencing Is Limited Only by Counting Statistics”, PLoS ONE, vol. 5, Issue 5 (e10439), May 3, 2010, 1-6.
Fan, J.-B. et al., “Highly Parallel SNP Genotyping”, Cold Spring Harbor Symposia on Quantitative Biology, vol. LXVIII, Feb. 2003, 69-78.
Fan, Jian-Bing et al., “Highly Parallel Genomic Assay”, Nature Reviews, 7, 2006, 632-644.
Fat Secret, “5 Foods to Never Eat”, https://www.fatsecret.com/calories-nutrition/food/white-bread/carboyhydrate (printed from internet Nov. 1, 2014)., 2 pages.
Fazio, Gennaro. et al., “Identification of RAPD Markers Linked to Fusarium Crown and Root Rot Resistance (Frl) in Tomato”, Euphytica 105, 1999, 205-210.
Fiorentino, F. et al., “Development and Clinical Application of a Strategy for Preimplantation Genetic Diagnosis of Single Gene Disorders Combined with HLA Matching”, Molecular Human Reproduction (Advance Access publication), 10 (6), 2004, 445-460.
Fiorentino, F et al., “Strategies and Clinical Outcome of 250 Cycles of Preimplantation Genetic Diagnosis for Single Gene Disorders”, Human Reproduction, 21, 3, 2006, 670-684.
Fiorentino, Francesco et al., “Short Tandem Repeats Haplotyping of the HLA Region in Preimplantation HLA Matching”, European Journal of Human Genetics, 13, 2005, 953-958.
Fitzgerald, “Intravascular Ultrasound Imaging of Coronary Arteries: Is Three Layers the Norm?”, Circulation, vol. 86, No. 1, Jul. 1, 1992, 154-158.
Ford, E. et al., “A method for generating highly multiplexed ChIP-seq libraries”, BMC Research Notes, vol. 7, No. 312, May 22, 2014, 1-5.
Forejt, et al., “Segmental trisomy of mouse chromosome 17: introducing an alternative model of Down's syndrome”, Genomics, 4(6), 2003, 647-652.
Forshew, et al., “Noninvasive Identification and Monitoring of Cancer Mutations by Targeted Deep Sequencing of Plasma DNA”, Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci. Transl. Med. 4, 136 30 (2012)., 1-12.
Forshew, T. et al., “Supplementary Materials for Noninvasive Identification and Monitoring of Cancer Mutations by Targeted Deep Sequencing of Plasma DNA”, Sci. Transl. Med, vol. 4, May 30, 2012, 20 pgs.
Fortina, P. et al., “Detection of the most common mutations causing beta-thalassemia in Mediterraneans using a multiplex amplification refractory mutation system (MARMS)”, Genome Res., vol. 2, 1992, 163-166.
Fortina, P. et al., “DOP-PCR Amplification of Whole Genomic DNA and Microchip-Based Capillary Electrophoresis”, Methods in Molecular Biology: Capillary Electrophoresis of Nucleic Acids, vol. II Practical Applications of Capillary Electrophoresis, 2001, 211-219.
Fu, G. K. et al., “Digital Encoding of Cellular mRNAs Enabling Precise and Absolute Gene Expression Measurement by Single-Molecule Counting”, Analytical Chemistry, vol. 86, Mar. 3, 2014, 2867-2870.
Fu, Yao-Wen et al., “Presence of Donor-and-recipientderived Dna Microchimerism in the Cell-free Blood Samples of Renal Transplantation Recipients Associates With The Acceptance of Transplanted Kidneys”, Asian Journal of Andrology, vol. 8, No. 4, Jul. 1, 2006, 477-482.
Gadi, V. K. et al., “Soluble Donor DNA Concentrations in Recipient Serum Correlate with Pancreas-Kidney Rejection”, Clinical Chemistry, vol. 52, No. 3, 2006, 379-382.
Ganshirt-Ahlert, D. et al., “Ratio of Fetal to Maternal DNA is Less Than 1 in 5000 at different Gestational Ages in Maternal Blood”, Clinical Genetics,38, 1990, 38-43.
Ganshirt-Ahlert, D. et al., “Fetal DNA in Uterine Vein Blood”, Obstetrics & Gynecology, 80 (4), 1992, 601-603.
Ganshirt-Ahlert, Dorothee et al., “Three Cases of 45,X/46,XYnf Mosaicism”, Human Genetics, 76, 1987, 153-156.
Gao, et al., “Relation of Donor Age and Preexisting Coronary Artery Disease on Angiography and Intracoronary Ultrasound to Later Development of Accelerated Allograft Coronary Artery Disease”, The American Journal of Cardiology, vol. 29, No. 3, Mar. 1, 1997, 623-629.
Gao, F. et al., “Characterizing Immunoglobulin Repertoire from Whole Blood by a Personal Genome Sequencer”, PLOS One, vol. 8, No. 9, Sep. 13, 2013, 8 pgs.
Gao, Ming et al., “Characterization of dull1, a Maize Gene Coding for a Novel Starch Synthase”, The Plant Cell, vol. 10, 1998, pp. 399-412.
Garcia-Murillas, I. et al., “Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer”, Science Translational Medicine, vol. 7, No. 302, Aug. 26, 2015, 34 pgs.
Gardina, P. et al., “Ploidy Status and Copy Number Aberrations in Primary Glioblastomas Defined by Integrated Analysis of Allelic Ratios, Signal Ratios and Loss of Heterozygosity Using 500K SNP Mapping Arrays”, BMC Genomics, 9 (489), (doi:10.1186/1471-2164-9-489), 2008, 16 pgs.
Gautier, E. et al., “Fetal RhD genotyping by maternal serum analysis: A two-year experience”, American Journal of Obstetrics and Gynecology, vol. 192, 2005, 666-669.
Geifman- Holtzman, et al., “Prenatal Diagnosis: Update on Invasive Versus Noninvasive Fetal Diagnostic Testing From Maternal Blood”, Expert Review of Molecular Diagnostics, vol. 8, No. 6, Nov. 1, 2008, 727-751.
Geiss, G. K. et al., “Direct multiplexed measurement of gene expression with color-coded probe pairs”, Nature Biotechnology, vol. 26, No. 3, Feb. 17, 2008, 317-325.
Ghanta, Sujana et al., “Non-Invasive Prenatal Detection of Trisomy 21 Using Tandem Single Nucleotide Polymorphisms”, PLoS ONE, 5 (10), 2010, 10 pgs.
Gholami, M. et al., “A tailed PCR procedure for cost-effective, two-order multiplex sequencing of candidate genes in polyploid plants”, Plant Biotechnology Journal, vol. 10, 2012, 635-645.
Gielis, E. M. et al., “Cell-Free DNA: An Upcoming Biomarker in Transplantation”, American Journal of Transplantation, vol. 15, 2015, 2541-2551.
Gielis, E. M. et al., “Plasma donor-derived cell-free DNA kinetics after kidney transplantation using a single tube multiplex PCR assay”, PLOS One, vol. 13, No. 12, e0208207, Dec. 6, 2018, 16 pgs.
Gineikiene, Egle et al., “Single Nucleotide Polymorphism-based System Improves the Applicability of Quantitative PCR for Chimerism Monitoring”, Journal of Molecular Diagnostics, vol. 11, No. 1, Jan. 1, 2009, 66-74.
Gingeras, et al., “Fifty Years of Molecular (DNA/RNA) Diagnostics”, Clinical Chemistry, vol. 51, No. 3, Jan. 13, 2005, 661-671.
Girnita, Diana M. et al., “Disparate Distribution of 16 Candidate Single Nucleotide Polymorphisms Among Racial and Ethnic Groups of Pediatric Heart Transplant Patients”, Transplantation, vol. 82, No. 12, Dec. 27, 2006, 1774-1780.
Gjertson, David W. et al., “Assessing Probability of Paternity and the Product Rule in DNA Systems”, Genetica, 96, 1995, 89-98.
Gnirke, A. et al., “Solution hybrid selection with ultra-long oligonucleotides for massively parallel targeted sequencing”, Nature Biotechnology, vol. 27, No. 2, Feb. 2009, 182-189.
Go, A. T. et al., “Non-invasive aneuploidy detection using free fetal DNA and RNA in maternal plasma: recent progress and future possibilities”, Human Reproduction Update, vol. 17, No. 3, 2011, 372-382.
Gordon, et al., “Disease-Specific Motifs Can Be Identified In Circulating Nucleic Acids From Live Elk and Cattle Infected With Transmissible Spongiform Encephalopathies”, Nucleic Acids Research, vol. 37. No. 2, Feb. 1, 2009, 550-556.
Gorringe, et al., “Large-scale Genomic Analysis of Ovarian Carcinomas”, Molecular oncology, vol. 3, No. 2, Apr. 1, 2009, 157-164.
Gouya, et al., “Coronary Artery Stenosis in High-risk Patients: 64-section Ct and Coronary Angiography—Prospective Study and Analysis of Discordance”, Radiology, vol. 252, No. 2, Aug. 1, 2009, 377-385.
Greenwalt, T. et al., “The Quantification of Fetomaternal Hemorrhage by an Enzyme-Linked Antibody Test with Glutaraldehyde Fixation”, Vox Sang, 63, 1992, 268-271.
Gregory, et al., “Comparison of Sixty-Four-Slice Multidetector Computed Tomographic Coronary Sngiography to Coronary Angiography With Intravascular Ultrasound for the Detection of Transplant Vasculopathy”, The American Journal of Cardiology, vol. 98, No. 7, Aug. 4, 2006, 877-884.
Griffiths, A. J. et al., “An Introduction to Genetic Analysis”, Sixth Edition, 1996, 5 pages.
Grskovic, M. et al., “Validation of a Clinical-Grade Assay to Measure Donor-Derived Cell-Free DNA in Solid Organ Transplant Recipients”, The Journal of Molecular Diagnostics, vol. 18, No. 6 + Supplemental Appendix S1, Nov. 2016, 890-902.
Grunenwald, H., “Optimization of Polymerase Chain Reactions”, Methods in Biology, vol. 226, 2003, 89-99.
Gu, H. et al., “Diagnostic role of microRNA expression profile in the serum of pregnant women with fetuses with neural tube defects”, Journal of Neurochemistry, vol. 122, 2012, 641-649.
Guerra, J., “Terminal Contributions for Duplex Oligonucleotide Thermodynamic Properties in the Context of Nearest Neighbor Models”, Biopolymers, 95(3), (2010), 2011, 194-201.
Guetta, Esther et al., “Analysis of Fetal Blood Cells in the Maternal Circulation: Challenges, Ongoing Efforts, and Potential Solutions”, Stem Cells and Development, 13, 2004, 93-99.
Guichoux, et al., “Current Trends in Microsatellite Genotyping”, Molecular Ecology Resources, 11, 2011, 591-911.
Gunderson, K. L. et al., “A genome-wide scalable SNP genotyping assay using microarray technology”, Nature Genetics, vol. 37, No. 5, May 2005, 549-554.
Gundry, C. N. et al., “Base-pair neutral homozygotes can be discriminated by calibrated high-resolution melting of small amplicons”, Nucleic Acids Research, vol. 36, No. 10, Apr. 29, 2008, 3401-3408.
Guo, H et al., “A Specific and Versatile Genome Walking Technique”, Gene, vol. 381, 2006, 18-23.
Gwee, Pai-Chung et al., “Simultaneous Genotyping of Seven Single-nucleotide Polymorphisms in the Mdr1 Gene by Single-tube Multiplex Minisequencing”, Pai-Chung Gwee. et al., “Simultaneous Genotyping of Seven Single-nucleotide Polymorphisms in the Mdr1 Gene by Single-tube Multiplex Minisequencing”, Clinical chemistry, Apr. 2003, vol. 49, Issue. 3, pp. 672-676., Apr. 1, 2003, 672-676.
Hahn, et al., “Non-invasive Prenatal Diagnostics Using Next Generation Sequencing: Technical, Legal and Social Challenges”, Expert Opinion on Medical Diagnostics, vol. 6, No. 6, Nov. 1, 2012, 517-528.
Hahn, S. et al., “Current applications of single-cell PCR”, CMLS Cellular and Molecular. Life Sciences, vol. 57, 2000, 96-105.
Hahn, S. et al., “Quantification of Circulating DNA: In the Preparation Lies the Rub”, Clinical Chemistry, vol. 47, No. 9, 2001, 1577-1578.
Halford, William P., “The Essential Prerequisites for Quantitative RT-PCR”, Nature Biotechnology, vol. 17, 1999, 1 page.
Hall, M., “Panorama Non-Invasive Prenatal Screening for Microdeletion Syndromes”, Apr. 1, 2014 (Apr. 1, 2014), XP055157224, Retrieved from the Internet: URL:http://www.panoramatest.com/sites/default/files/files/PanoramaMicrodeletionsWhite Paper-2.pdf [retrieved on Dec. 8, 2014].
Han, S-W et al., “Predictive and Prognostic Impact of Epidermal Growth Factor Receptor Mutation in Non-Small-Cell Lung Cancer Patients Treated With Gefitinib”, Journal of Clinical Oncology, vol. 23, No. 11, Apr. 10, 2005, 2493-2501.
Handley, D. et al., “Noninvasive prenatal chromosomal aneuploidy detection using plasma cell-free nucleic acid”, Expert Rev Obstet. Gynecol, vol. 5, No. 5, 2010, 581-590.
Handyside, et al., “Isothermal whole genome amplification from single and small numbers of cells: a new era for preimplantation genetic diagnosis of inherited disease”, Molecular Human Reproduction vol. IO, No. 10 pp. 767-772, 2004.
Hao, T. B. et al., “Circulating cell-free DNA in serum as a biomarker for diagnosis and prognostic prediction of colorectal cancer”, British Journal of Cancer, vol. 111, Aug. 26, 2014, 1482-1489.
Hara, Eiji et al., “Subtractive eDNA cloning using oligo(dT)30-latex and PCR: isolation of eDNA clones specific to undifferentiated human embryonal carcinoma cells”, Nucleic Acids Research, 19(25), 1991, 7097-7104.
Hardenbol, P., “Multiplexed Genotyping With Sequence-Tagged Molecular Inversion Probes”, Nature Biotechnology, 21 (6), 2003, 673-678.
Hardenbol, Paul et al., “Highly multiplexed molecular inversion probe genotyping: Over 10,000 targeted SNPs genotyped in a singled tube assay”, Genome Research, 15, 2005, 269-275.
Harismendy, O. et al., “Method for Improving Sequence Coverage Uniformity of Targeted Genomic Intervals Amplified by LR-PCR Using Illumina GA Sequencing-By-Synthesis Technology”, Bio Techniques, 46(3), 2009, 229-231.
Harper, J. C. et al., “Recent Advances and Future Developments in PGD”, Prenatal Diagnosis, 19, 1999, 1193-1199.
Harton, G.L. et al., “Preimplantation Genetic Testing for Marfan Syndrome”, Molecular Human Reproduction, 2 (9), 1996, 713-715.
Hartwell, L. H. et al., “Chapter 11: The Direct Detection of Genotype Distinguishes Individual Genomes”, Genetics: From Genes to Genomes, Second Edition, 2004, 371-414.
Hartwell, L. H. et al., “Chapter 13: Chromosomal Rearrangements and Changes in Chromosome Number Reshape Eukaryotic Genomes”, Genetics: From Genes to Genomes, Second Edition, 2004, 441-486.
Hattori, M. et al., “The DNA sequence of human chromosome 21”, Nature, vol. 405, May 18, 2000, 311-319.
Hawkins, T. et al., “Whole genome amplification—applications and advances”, Current Opinion in Biotechnology, 13, 2002, 65-67.
Hayden, et al., “Multiplex-Ready PCR: A new method for multiplexed SSR and SNP genotyping”, BMC Genomics 2008, 9(80), 1-12.
He, QZ et al., “A method for improving the accuracy of non-invasive prenatal screening by cell-free foetal DNA size selection”, British Journal of Biomedical science, vol. 75, No. 3, Jul. 2018, 133-138.
Heaton, Paul R. et al., “Heminested PCR Assay for Detection of Six Genotypes of Rabies and Rabies-related Viruses”, Journal of Clinical Microbiology, vol. 35, 1997, pp. 2762-2766.
Heidary, M. et al., “The dynamic range of circulating tumor DNA in metastatic breast cancer”, Breast Cancer Research, vol. 16, No. 421, 2014, 10 pages.
Hellani, A. et al., “Clinical Application of Multiple Displacement Amplification in Preimplantation Genetic Diagnosis”, Reproductive BioMedicine Online, 10 (3), 2005, 376-380.
Hellani, Ali et al., “Multiple displacement amplification on single cell and possible PGD applications”, Molecular Human Reproduction, 10(11), 2004, 847-852.
Henegariu, O. et al., “Multiplex PCR: Critical Parameters and Step-by-Step Protocol”, Biotechniques, vol. 23, 1997, pp. 504-511.
Hidestrand, M. et al., “Highly Sensitive Noninvasive Cardiac Transplant Rejection Monitoring Using Targeted Quantification of Donor-Specific Cell-Free Deoxyribonucleic Acid”, Journal of the American College of Cardiology, vol. 63, No. 12, 1224-1226, 2014.
Hoberman, Rose et al., “A Probabilistic Approach for SNP Discovery in High-throughput Human Resequencing Data”, Genome Research, vol. 19, Jul. 15, 2009, 1542-1552.
Hochberg, et al., “A Novel Rapid Single Nucleotide Polymorphism (SNP)-Based Method for Assessment of Hematopoietic Chimerism After Allogeneic Stem Cell Transplantation”, Blood, vol. 101, No. 1, Jan. 1, 2003, 363-369.
Hodges, et al., “Genome-wide In Situ Exon Capture for Selective Resequencing”, Nature Genetics, vol. 39, No. 12, Nov. 4, 2007, 1522-1527.
Hodgkinson, C. L. et al., “Tumorigenicity and genetic profiling of circulating tumor cells in small-cell lung cancer”, Nature Medicine, vol. 20, No. 8, Aug. 2014, 897-905.
Hoffmann, Steven et al., “Donor Genomics Influence Graft Events: The Effect of Donor Polymorphisms on Acute Rejection and Chronic Allograft Nephropathy”, Kidney International, vol. 66, No. 4, Oct. 1, 2004, 1686-1693.
Hojsgaard, S. et al., “BIFROST—Block recursive models induced from relevant knowledge, observations, and statistical techniques”, Computational Statistics & Data Analysis, 19(2), 1995, 155-175.
Hollas, B. et al., “A stochastic approach to count RN A molecules using DNA sequencing methods”, Lecture Notes in Computer Science, vol. 2812, 2003, 55-62.
Holleley, et al., “Multiplex Manager 1.0: a Cross-Platform Computer Program that Plans and Optimizes Multiplex PCR”, BioTechniques46:511-517 (Jun. 2009), 511-517.
Hollox, E. et al., “Extensive Normal Copy Number Variation of a ß-Defensin Antimicrobial-Gene Cluster”, Am. J. Hum. Genet., 73, 2003, 591-600.
Holt, et al., “Detecting SNPS and Estimating Allele Frequencies in Clonal Bacterial Populations by Sequencing Pooled DNA”, Bioinformatics, vol. 25, No. 16, Aug. 15, 2009, 2074-2075.
Homer, et al., “Resolving Individuals Contributing Trace Amounts of DNA to Highly Complex Mixtures Using High-Density SNP Genotyping Microarrays”, PLOS Genetics, 4(8), 2008, 9 pgs.
Hoogendoorn, Bastiaan et al., “Genotyping Single Nucleotide Polymorphisms by Primer Extension and High Performance Liquid Chromatography”, Hum Genet, 104, 1999, 89-93.
Horai, et al., “Novel Implantable Device to Detect Cardiac Allograft Rejection”, Circulation, vol. 120, No. Suppl 1, Sep. 15, 2009, 185-190.
Hornak, M. et al., “Aneuploidy Detection in Pigs Using Comparative Genomic Hybridization: From the Oocytes to Blastocysts”, PLoS ONE, vol. 7, No. 1, Jan. 2012, 6 pages.
Hosmillo, Myra D. et al., “Development of Universal SYBR Green Real-time RT-PCR for the Rapid Detection and Quantitation of Bovine and Porcine Toroviruses”, Journal of Virological Methods, vol. 168, 2010, pp. 212-217.
Hosono, S. et al., “Unbiased Whole-Genome Amplification Directly From Clinical Samples”, Genome Research, vol. 13, 2003, 954-964.
Hospital, F. et al., “A General Algorithm to Compute Multilocus Genotype Frequencies Under Various Mating Systems” vol. 12, No. 6, Jan. 1, 1996 (Jan. 1, 1996), pp. 455-462.
Hou, X. et al., “Analysis of the Repertoire Features of TCR Beta Chain CDR3 in Human by High-Throughput Sequencing”, Cellular Physiology and Biochemistry, vol. 39, Jul. 21, 2019, 651-667.
Howie, et al., “Fast and accurate genotype imputation in genome-wide association studies through pre-phasing”, Nature Genetics, vol. 44, No. 8, Jul. 22, 2012, 955-959.
Howie, B. N. et al., “A Flexible and Accurate Genotype Imputation Method for the Next Generation of Genome-Wide Association Studies”, PLoS Genetics, vol. 5, No. 6, Jun. 2009, 15 pages.
Hu, Dong Gui et al., “Aneuploidy Detection in Single Cells Using DNA Array-Based Comparative Genomic Hybridization”, Molecular Human Reproduction, 10(4), 2004, 283-289.
Hu, Hao et al., “Mutation Screening in 86 Known X-linked Mental Retardation Genes by Droplet-based Multiplex Pcr and Massive Parallel Sequencing”, Hao Hu. et al., “Mutation Screening in 86 Known X-linked Mental Retardation Genes by Droplet-based Multiplex Pcr and Massive Parallel Sequencing”, Hugo J, Dec. 2009, vol. 3, pp. 41-49., Dec. 1, 2009, 41-49.
Hu, Y. et al., “Detection of Extrahepatic Hepatitis C Virus Replication by a Novel, Highly Sensitive, Single-Tube Nested Polymerase Chain Reaction”, Am. J. Clin Pathol., vol. 119, 2003, 95-100.
Huang, D. J. et al., “Reliable detection of Trisomy 21 using MALDI-TOF mass spectrometry”, Genetics in Medicine, vol. 8, Nov. 2006, 728-734.
Huang, D. J. et al., “Use of an Automated Method Improves the Yield and Quality of Cell-Free Fetal DNA Extracted from Maternal Plasma”, Clinical Chemistry, vol. 51, No. 12, 2005, 2419-2420.
Huang, J. et al., “Whole genome DNA copy number changes identified by high density oligonucleotide arrays”, Human Genomics, vol. 1, No. 4, May 2004, 287-299.
Hubacek, et al., “Detection of Donor DNA After Heart Transplantation: How Far Could It Be Affected by Blood Transfusion and Donor Chimerism?”, Transplantation Proceedings, vol. 39, Jun. 1, 2007, 1593-1595.
Hug, H. et al., “Measurement of the No. of molecules of a single mRNA species in a complex mRNA preparation”, J. Theor. Biol., vol. 221, 2003, 615-624.
Hultin, E. et al., “Competitive enzymatic reaction to control allele-specific extensions”, Nucleic Acids Research, vol. 33, No. 5, Mar. 14, 2005, 1-10.
Hung, E.C.W. et al., “Detection of circulating fetal nucleic acids: a review of methods and applications”, J. Clin. Pathol., vol. 62, 2009, 308-313.
Hyndman, D L. et al., “PCR Primer Design”, Methods in Molecular Biology, vol. 226, Second Edition, 2003, 81-88.
Ido, Yasuo et al., “Hyperglycemia-Induced Apoptosis in Human Umbilical Vein Endothelial Cells: Inhibition by the AMP-Activated Protein Kinase Activation”, Diabetes, 51, 2002, 159-167.
Illumina, “Automated GoldenGate™ Genotyping on the BeadStation 500”, Pub. No. 970-2004-002, 2004, 2 pages.
Illumina, “Genomic Sequencing”, Data Sheet: Sequencing, 2010, 38939-38944.
Illumina, “GoldenGate” Assay Workflow: Illumina's GoldenGate assay protocol provides high-quality, high-multiplex genotyping results with a streamlined workflow, Pub. No. 370-2004-006, 2004, 2 pages.
Illumina, “HiSeq 2500 Sequencing System”, System Specification Sheet: Sequencing, available via URL https://www.illumina.com/documents/products/datasheets/datasheet_hiseq2500.pdf, 2015, 4 pgs.
Illumina, “History of Sequencing by Synthesis”, https://www.illumina.com/science/technology/next-generation-sequencing/illumina-sequencing-history.html, 2020, 3 pages.
Illumina, “Illumina Extends BeadArray Technology to Address Wider Range of SNP Genotyping Projects; New Microarray Offerings Enable Genotyping at 384 and 786 Multiplex”, Businesswire, May 4, 2004, 2 pages.
Illumina, “Illumina® Beadstation 500: A Scalable System That Grows With Your Research Requirements”, Pub. No. 970-2005-003, Jul. 1, 2005, 4 pages.
Illumina, “Illumina Announces Benchtop SNP Genotyping System”, Press Release, Nov. 5, 2003, 3 pages.
Illumina, “Illumina Begins Shipment of BeadStation 500G Benchtop Genotyping System”, Press Release, Apr. 15, 2004, 3 pages.
Illumina, “MiSeq System Information Sheet”, 2018, 3 pgs.
Illumina, “Patent Owner Illumina's Preliminary Response to Petition”, Oct. 17, 2018, 75 pgs.
Illumina, “Petition for Inter Partes Review of U.S. Pat. No. 8,682,592”, Jun. 13, 2019, 91 pages.
Illumina, “Plaintiff/Counterclaim Defendant Illumina, Inc.'s Amended Patent L.R. 3-3 Preliminary Invalidity Contentions for U.S. Pat. No. 8,682,592”, Oct. 30, 2018, 22 pages.
Illumina, “Plaintiff/Counterclaim-Defendant Illumina, Inc.'s Patent L.R. 3-3 Contentions for U.S. Patent Preliminary Invalidity Contentions for U.S. Pat. No. 8,682,592”, Oct. 9, 2018, 81 pages.
Illumina, “Preparing Samples for Sequencing Genomic DNA”, Part # 11251892 Rev. A, 2007, 18 pages.
Illumina, “Preparing Samples for Sequencing Genomic DNA”, Part # 1003806 Rev. A, 2007, 20 pages.
Illumina, “Products & Services”, Product Literature, Mar. 21, 2007, 3 pages.
Illumina, “Technology: Solexa Sequencing Technology”, May 21, 2007, 1 page.
Illumina, “TruSeq™ RNA and DNA Library Preparation Kits v2”, Data Sheet: Illumina® Sequencing, 2014, 4.
Illumina Catalog, “Paired-End Sample Preparation Guide, Illumina Catalog# PE-930-1 001, Part# 1005063 Rev. E”, 2011, 1-40.
Illumina, Inc., “Declaration of David Peters, Ph.D. in Support of Petition for Inter Partes Review of U.S. Pat. No. 8,682,592”, Jun. 13, 2019, 136 pages.
Illumina, Inc. v. Natera, Inc., “Order Re: Claim Construction”, Jan. 30, 2019, 16 pgs.
Imielinski, M. et al., “Mapping the Hallmarks of Lung Adenocarcinoma with Massively Parallel Sequencing”, Cell, vol. 150, Sep. 14, 2012, 1107-1120.
Ingman, et al., “SNP Frequency Estimation Using Massively Parallel Sequencing of Pooled DNA”, European Journal of Human Genetics, vol. 17, No. 3, Oct. 15, 2008, 383-386.
Innan, H. et al., “The Pattern of Polymorphism on Human Chromosome 21”, Genome Research, vol. 13, 2003, 1158-1168.
Interewicz, B. et al., “DNA Released from Ischemic and Rejecting Organs as an Indicator of Graft Cellular Damage”, Annals of Transplantation, vol. 9, No. 2, May 1, 2004, 42-45.
International Human, Genome Sequencing Consortium , “Finishing the Euchromatic Sequence of the Human Genome”, Nature, vol. 431, Oct. 21, 2004, 931-945.
Ishii, et al., “Optimization of Annealing Temperature to Reduce Bias Caused by a Primer Mismatch in Multitemplate PCR”, Applied and Environmental Microbiology, Aug. 2001, p. 3753-3755.
Iskow, R. C. et al., “Natural Mutagenesis of Human Genomes by Endogenous Retrotransposons”, Cell, vol. 141, Jun. 25, 2010, 1253-1261.
Ivanov, M. et al., “Non-random fragmentation patterns in circulating cell-free DNA reflect epigenetic regulation”, BMC Genomics, vol. 16 (Suppl 13):S1, Jun. 2015, 12 pgs.
Jabara, C. B. et al., “Accurate sampling and deep sequencing of the HIV-1 protease gene using a Primer ID”, PNAS, vol. 108, No. 50, Dec. 13, 2011, 20166-20171.
Jahr, S. et al., “DNA Fragments in the Blood Plasma of Cancer Patients: Quantitations and Evidence for Their Origin from Apoptotic and Necrotic Cells”, Cancer Research, vol. 61, Feb. 15, 2001, 1659-1665.
Jamal-Hanjani, M. et al., “Detection of ubiquitous and heterogeneous mutations in cell-free DNA from patients with early-stage non-small-cell lung cancer”, Annals of Oncology, vol. 27, No. 5, Jan. 28, 2016, 862-867.
Jamal-Hanjani, M. et al., “Tracking Genomic Cancer Evolution for Precision Medicine: The Lung TRACERx Study”, PLOS Biology, vol. 12, No. 7, Jul. 2014, 1-7.
Jamal-Hanjani, M. et al., “Tracking the Evolution of Non-Small-Cell Lung Cancer”, The New England Journal of Medicine, vol. 376, No. 22, Jun. 1, 2017, 2109-2121.
Jarvie, T., “Next generation sequencing technologies”, Drug Discovery Today: Technologies, vol. 2, No. 3, 2005, 255-260.
Jen, J. et al., “An Overview on the Isolation and Analysis of Circulating Tumor DNA in Plasma and Serum”, Annals New York Academy of Sciences, 2000, 8-12.
Jenkins, S. et al., “High-throughput SNP genotyping”, Comparative and Functional Genomics, vol. 3, Dec. 5, 2001, 57-66.
Jennings, C. et al., “Investigation of Effects of Acid Citrate Dextrose and EDTA on Ability to Quantitatively Culture Human Immunodeficiency Virus”, Journal of Clinical Microbiology, vol. 38, No. 9, 2000, 3522.
Jett, K. et al., “Clinical and genetic aspects of neurofibromatosis 1”, Genetics in Medicine, vol. 12, No. 1, Jan. 2010, 11 pages.
Jewesburty, E.C.O., “Reactions after Transfusion of Stored Blood”, The British Medical Journal, vol. 1, No. 4191, 1941, 664-665.
Jiang, P. et al., “The Long and Short of Circulating Cell-Free DNA and the Ins and Outs of Molecular Diagnostics”, Trends in Genetics, vol. 32, No. 6, Jun. 2016, 360-371.
Johnson, D. S. et al., “Genome-Wide Mapping of in Vivo Protein-DNA Interactions”, Science, vol. 316, Jun. 8, 2007, 1497-1502.
Johnson, D.S. et al., “Comprehensive Analysis of Karyotypic Mosaicism Between Trophectoderm and Inner Cell Mass”, Molecular Human Reproduction, 16(12), 2010, 944-949.
Johnson, J. B. et al., “Differential mechanisms of complementmediated neutralization of the closely related paramyxoviruses simian virus 5 and mumps virus”, Virology, vol. 376, No. 1, 2008, 112-123.
Johnson, K. L. et al., “Interlaboratory Comparison of Fetal Male DNA Detection from Common Maternal Plasma Samples by Real-Time PC”, Clinical Chemistry, vol. 50, No. 3, 2004, 516-521.
Johnson D.S, et al., “Preclinical Validation of a Microarray Method for Full Molecular Karyotyping of Blastomeres in a 24-h Protocol”, Human Reproduction, 25 (4), 2010, 1066-1075.
Jung, K. et al., “Cell-free DNA in the blood as a solid tu1nor biomarker-A critical appraisal of the literature”, Clinica Chimica Acta, vol. 411, 2010, 1611-1624.
Juppner, H. et al., “Functional Properties of the PTH/PTHrP Receptor”, Bone, vol. 17, No. 2 Supplement, Aug. 1995, 39S-42S.
Kalendar, Ruslan et al., “Java Web Tools for PCR, in Silico PCR, and Oligonucleotide Assembly and Analysis”, Genomics, vol. 98, 2011, pp. 137-144.
Kamat, A. A. et al., “Quantification of total plasma cell-free DNA in ovarian cancer using real-time PCR”, Ann N Y Acad Sci., vol. 1075, Sep. 2006, 230-234.
Kamel, A. M. et al., “A simple strategy for breakpoint fragment determination in chronic myeloid leukemia”, Cancer Genetics and Cytogenetics, vol. 122, 2000, 110-115.
Kane, M. et al., “Application of Less Primer Method to Commercial Kits”, Forensic Science International: Genetics Supplement Series, vol. 1, Issue 1, 2008, 41-43.
Kane, M., “Application of Less Primer Method to Multiplex PCR”, International Congress Series, vol. 1288, 2006, pp. 694-696.
Kapadia, Samir R. et al., “Impact of Intravascular Ultrasound in Understanding Transplant Coronary Artery Disease”, Current Opinion in Cardiology, vol. 14, No. 2, Mar. 1, 1999, 1-19.
Kaplinski, Lauris et al., “MultiPLX: Automatic Grouping and Evaluation of PCR Primers”, Bioinformatics, 21(8), 2005, 1701-1702.
Karger, et al., “DNA Sequencing by Capillary Electrophoresis”, Electrophoresis, vol. 30, Supplement 1, Jun. 1, 2009, 1-11.
Karoui, Noureddine E. et al., “Getting More from Digital SNP Data”, Statistics in Medicine, vol. 25, Issue 18, Jan. 5, 2006, 3124-3133.
Kass, et al., “Diagnosis of Graft Coronary Artery Disease”, Current Opinion in Cardiology, vol. 22, No. 2, Mar. 1, 2007, 139-145.
Kathiresan, Sekar et al., “Genome-wide Association of Early-onset Myocardial Infarction With Common Single Nucleotide Polymorphisms, Common Copy Number Variants, and Rare Copy Number Variants”, Nature Genetics, vol. 41, No. 3, Mar. 1, 2009, 1-23.
Kazakov, V.I. et al., “Extracellular DNA in the Blood of Pregnant Women”, Tsitologia, vol. 37, No. 3, 1995, 1-8.
Keith, L. et al., “Clinical Experience With the Prevention of Rh-Isoimmunization: A Historical Comparative Analysis”, American Journal of Reproductive Immunology, vol. 5, 1984, 84-89.
Keller, M. C. et al., “Non-Pathological Paternal Isodisomy of Chromosome 2 Detected From a Genome-Wide SNP Scan”, American Journal of Medical Genetics, Part A, 2009, 1823-1826.
Kennedy, S. R. et al., “Detecting ultralow-frequency mutations by Duplex Sequencing”, Nature Protocols, vol. 9, No. 11, 2014, 2586-2606.
Kibbe, Warren A., “Oligocalc: An Online Oligonucleotide Properties Calculator”, Nucleic Acids Research, vol. 35, 2007, pp. W43-W46.
Kiernan, J. A., “Formaldehyde, formalin, paraformaldehyde and glutaraldehyde: What they are and what they do.”, Microscopy Today, vol. 1, 2000, 8-12.
Kijak, G. et al., “Discrepant Results in the Interpretation of HIV-1 Drug-Resistance Genotypic Data Among Widely Used Algorithms”, HIV Medicine, 4, 2003, 72-78.
Kim, H. et al., “Whole-genome and multisector exome sequencing of primary and post-treatment glioblastoma reveals patterns of tumor evolution”, Genome Research, vol. 25, No. 3, Feb. 3, 2015, 316-327.
Kimmel, G. et al., “GERBIL: Genotype resolution and block identification using likelihood”, PNAS, vol. 102, No. 1, Jan. 4, 2005, 158-162.
Kinde, I. et al., “Detection and quantification of rare mutations with massively parallel sequencing”, PNAS, vol. 108, No. 23, Jun. 7, 2011, 9530-9535.
Kinnings, S. L. et al., “Factors affecting levels of circulating cell-free fetal DNA in maternal plasma and their implications for noninvasive prenatal testing”, Prenatal Diagnosis, vol. 35, 2015, 816-822.
Kircher, Martin et al., “Improved Base Calling for the Illumina Genome Analyzer Using Machine Learning Strategies”, Genome Biology, vol. 10, Issue 8, Article No. R83, Aug. 14, 2009, 83.2-83.9.
Kirkizlar, E. et al., “Detection of Clonal and Subclonal Copy-Number Variants in Cell-Free DNA from Patients with Breast Cancer Using a Massively Multiplexed PCR Methodology”, Translational Oncology, vol. 8, No. 5, Oct. 2015, pp. 407-416.
Kirkness, E. F. et al., “Sequencing of isolated sperm cells for direct haplotyping of a human genome”, Genome Research, vol. 23, 2013, 826-832.
Kivioja, T. et al., “Counting absolute number of molecules using unique molecular identifiers”, Nature Proceedings, Apr. 14, 2011, 18 pgs.
Kivioja, T. et al., “Counting absolute numbers of molecules using unique molecular identifiers”, Nature Methods, Advance Online Publication, Nov. 20, 2011, 1-5.
Kivioja, T. et al., “Counting absolute numbers of molecules using unique molecular identifiers”, Nature Methods, vol. 9, No. 1, Jan. 2012, 72-76.
Kobashigawa, et al., “Multicenter Intravascular Ultrasound Validation Study Among Heart Transplant Recipients”, Journal of the American College of Cardiology, vol. 45, No. 9, May 3, 2005, 1532-1537.
Koboldt, et al., “VarScan: Variant Detection In Massively Parallel Sequencing of Individual and Pooled Samples”, Bioinformatics, vol. 25, No. 17, Jun. 19, 2009, 2283-2285.
Koelman, et al., “Donor-derived Soluble HLA Plasma Levels Can Not Be Used to Monitor Graft Rejection in Heart Transplant Recipients”, Transplant Immunology, vol. 8, No. 1, Mar. 1, 2000, 57-64.
Kohler, C. et al., “Levels of plasma circulating cell free nuclear and mitochondrial DNA as potential biomarkers for breast tumors”, Molecular Cancer, vol. 8, No. 105, Nov. 17, 2009, 9 pages.
Koide, K. et al., “Fragmentation of cell-free fetal DNA in plasma and urine of pregnant women”, Prenatal Diagnosis, vol. 25, 2005, 604-607.
Koldehoff, Michael et al., “Quantitative analysis of chimerism after allogeneic stem cell transplantation by real-time polymerase chain reaction with single nucleotide polymorphisms, standard tandem repeats, and Y-chromosome-specific sequences”, American Journal of Hematology, vol. 81, No. 10, Jul. 12, 2006, 735-746.
Konfortov, B A. et al., “A High-Resolution HAPPY Map of Dictyostelium discoideum Chromosome 6”, Genome Research, vol. 10, No. 11, Nov. 2000, 1737-1742.
Konfortov, Bernard A. et al., “An Efficient Method for Multi-Locus Molecular Haplotyping”, Nucleic Acids Research, 35(1), e6, 2007, 8 pgs.
Kopreski, MS et al., “Detection of mutant K-ras DNA in plasma or serum of patients with colorectal cancer”, British Journal of Cancer, vol. 76, No. 10, 1997, 1293-1299.
Koressaar, Triinu et al., “Enhancements and Modifications of Primer Design Program Primer3”, Bioinformatics, vol. 23, 2007, pp. 1289-1291.
Korn, et al., “Integrated Genotype Calling and Association Analysis of SNPS, Common Copy Number Polymorphisms and Rare CNVS”, Nature Genetics, vol. 40, No. 10, Oct. 1, 2008, 1253-1260.
Krjutskov, K. et al., “Development of a single tube 640-plex genotyping method for detection of nucleic acid variations on microarrays”, Nucleic Acids Research, vol. 36, No. 12, May 23, 2008, 7 pages.
Kuhn, H. et al., “Rolling-circle amplification under topological constraints”, Nucleic Acids Research, vol. 30, No. 2, 2002, 574-580.
Kukita, Y. et al., “High-fidelity target sequencing of individual molecules identified using barcode sequences: de nova detection and absolute quantitation of mutations in plasma cell-free DNA from cancer patients”, DNA Research, vol. 22, No. 4, Jun. 29, 2015, 269-277.
Kuliev, Anver et al., “Thirteen Years' Experience on Preimplantation Diagnosis: Report of the Fifth International Symposium on Preimplantation Genetics”, Reproductive BioMedicine Online, 8, 2, 2004, 229-235.
Kumar, P. et al., “Ethylenegycol-Bis-(B-Aminoethylether)Tetraacetate as a Blood Anticoagulant: Preservation of Antigen-Presenting Cell Function and Antigen-Specific Proliferative Response of Peripheral Blood Mononuclear Cells from Stored Blood”, Clinical and Diagnostic Laboratory Immunology, vol. 7, No. 4, 2000, 578-583.
Kunishima, S. et al., “First description of somatic mosaicism in MYH9 disorders”, British Journal of Haematology, vol. 128, 2005, 360-365.
Kwok, P. Y., “High-throughput genotyping assay approaches”, Pharmacogenomics, vol. 1, No. 1, 2000, 1-5.
Lambert, et al., “Quantification of Maternal Microchimerism by HLA-Specific Real-time Polymerase Chain Reaction”, Arthritis and Rheumatism, vol. 50, No. 3, Mar. 1, 2004, 906-914.
Lambert-Messerlian, G. et al., “Adjustment of Serum Markers in First Trimester Screening”, Journal of Medical Screening, 16 (2), 2009, 102-103.
Landegren, U. et al., “Padlock and proximity probes for in situ and array-based analyses: tools for the post-genomic era”, Comparative and Functional Genomics, vol. 4, 2003, 525-530.
Lander, E. S. et al., “Initial sequencing and analysis of the human genome”, Nature, vol. 409, Feb. 15, 2001, 860-921.
Langmore, J. , “Quality Control and Pre-Qualifications of NGS Libraries Made from Clinical Samples”, ABRF 2013 Satellite Workshop, Mar. 2, 2013, 35 pages.
Lanman, et al., “Analytical and Clinical Validation of a Digital Sequencing Panel for Quantitative, Highly Accurate Evaluation of Cell-Free Circulating Tumor DNA”, Plos One, DOI:10.1371/journal.pone.0140712, 2015, 1-27.
Lapaire, O. et al., “Array-CGH analysis of cell-free fetal DNA in 10 mL of amniotic fluid supernatant”, Prenatal Diagnosis, vol. 27, May 17, 2007, 616-621.
Lapierre, J.M. et al., “Analysis of uncultured amniocytes by comparative genomic hybridization: a prospective prenatal study”, Prenatal Diagnosis, vol. 20, 2000, 123-131.
Lardeux, Frederic et al., “Optimization of a Semi-nested Multiplex PCR to Identify Plasmodium Parasites in Wild-Caught Anopheles in Bolivia, and Its Application to Field Epidemiological Studies”, Transactions of the Royal Society of Tropical Medicine and Hygiene, vol. 102, 2008, pp. 485-492.
Larsen, J. B. et al., “Single-step Nested Multiplex PCR to Differentiate Between Various Bivalve Larvae”, Marine Biology, vol. 146, 2005, pp. 1119-1129.
Lasken, R. S. et al., “Whole genome amplification: abundant supplies of DNA from precious samples or clinical specimens”, Trends in Biotechnology, vol. 21, No. 12, Dec. 2003, 531-535.
Lathi, Ruth B. et al., “Informatics Enhanced SNP Microarray Analysis of 30 Miscarriage Samples Compared to Routine Cytogenetics”, PLoS ONE, 7(3), 2012, 5 pgs.
Lavebrat, et al., “Single Nucleotide Polymorphism (SNP) Allele Frequency Estimation in DNA Pools Using Pyrosequencing”, Nature Protocols, vol. 1, No. 6, Jan. 11, 2007, 2573-2582.
Lavebratt, Catharina et al., “Pyrosequencing-based SNP Allele Frequency Estimation in DNA Pools”, Human Mutation, vol. 23, Issue 1, Dec. 19, 2003, 92-97.
Lavrentieva, I et al., “High Polymorphism Level of Genomic Sequences Flanking Insertion Sites of Human Endogenous Retroviral Long Terminal Repeats”, FEBS Letters, vol. 443, No. 3, Jan. 29, 1999, 341-347.
Leamon, John H. et al., “A massively parallel Pico TiterPlate based platform for discrete picoliter-scale polymerase chain reactions”, Electrophoresis, vol. 24, No. 21, Nov. 1, 2003, 3769-3777.
Leary, R. J. et al., “Development of Personalized Tumor Biomarkers Using Massively Parallel Sequencing”, Science Translational Medicine, vol. 2, No. 20, Feb. 24, 2010, 1-8.
Leary, Rebecca J et al., “Detection of Chromosomal Alterations in the Circulation of Cancer Patients with Whole-Genome Sequencing”, Science Translational Medicine, 4, 162, 2012, 12.
Lecomte, T. et al., “Detection of Free-Circulating Tumor-Associated DNA in Plasma of Colorectal Cancer Patients and Its Association With Prognosis”, Int. J. Cancer, vol. 100, 2002, 542-548.
Lee, et al., “ERBB2 kinase domain mutation in the lung squamous cell carcinoma”, Cancer Letters, vol. 237, 2006, 89-94.
Lee, J et al., “Anchored Multiplex PCR Enables Sensitive and Specific Detection of Variants in Circulating Tumor DNA by Next-Generation Sequencing”, DOI:https://doi.org/10.1016/j.cancergen.2017.04.049, Cancer Genetics 214-215, 2017, 47.
Lee, T. et al., “Down syndrome and cell-free fetal DNA in archived maternal serum”, AmJ Obstet Gynecol, vol. 187, No. 5, 1217-1221, Nov. 2002.
Lee, T.H. et al., “Quantitation of genomic DNA in plasma and serum samples: higher concentrations of genomic DNA found in serum than in plasma”, Transfusion, vol. 41, Feb. 2001, 276-282.
Levsky, J. M. et al., “Fluorescence in situ hybridization: past, present and future”, Journal of Cell Science, vol. 116, No. 14, 2003, 2833-2838.
Levsky, Jeffrey M. et al., “Efficacy of Coronary Ct Angiography: Where We Are, Where We Are Going and Where We Want to Be”, Journal of Cardiovascular Computed Tomography, vol. 3, Supplement 2, Nov. 2, 2009, s99-s108.
Li, et al., “Detection of SNPs in the Plasma of Pregnant Women and in the Urine of Kidney Transplant Recipients by Mass Spectrometry”, Annals of the New York Academy of Sciences, vol. 1075, Sep. 5, 2006, 144-147.
Li, et al., “Mapping Short DNA Sequencing Reads and Calling Variants Using Mapping Quality Scores”, Genome Research, vol. 18, No. 11,, Aug. 19, 2008, 1851-1858.
Li, et al., “Multiplex Padlock Targeted Sequencing Reveals Human Hypermutable CpG Variations”, Genome Research, vol. 19, No. 9, Jun. 12, 2009, 1606-1615.
Li, et al., “SOAP2: An Improved Ultrafast Tool for Short Read Alignment”, Bioinformatics, vol. 25, No. 15, Aug. 1, 2009, 1966-1967.
Li, B., “Highly Multiplexed Amplicon Preparation for Targeted Re-Sequencing of Sample Limited Specimens Using the Ion AmpliSeq Technology and Semiconductor Sequencing”, Proceedings of the Annual Meeting of the American Society of Human Genetics [retrieved on Oct. 30, 2012]. Retrieved from the Internet: <URL: http://www.ashg.org/2012meeting/abstracts/fulltext/f120121811.htm>, 2012, 1 pg.
Li, R. et al., “SNP detection for massively parallel whole-genome resequencing”, Genome Research, vol. 19, 2009, 1124-1132.
Li, Y. et al., “Detection of Paternally Inherited Fetal Point Mutations for b-Thalassemia Using Size-Fractionated Cell-Free DNA in Maternal Plasma”, JAMA, vol. 293, No. 7, Apr. 13, 2005, 843-849.
Li, Y. et al., “Non-Invasive Prenatal Diagnosis Using Cell-Free Fetal DNA in Maternal Plasma from PGD Pregnancies”, Reproductive BioMedicine Online, 19 (5), 2009, 714-720.
Li, Ying et al., “Detection of Donor-specific DNA Polymorphisms in the Urine of Renal Transplant Recipients”, Clinical Chemistry, vol. 49, No. 4, Apr. 1, 2003, 655-658.
Li, Ying et al., “Ready detection of donor-specific single-nucleotide polymorphisms in the urine of renal transplant recipients by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry”, Ying Li. et al., “Ready detection of donor-specific single-nucleotide polymorphisms in the urine of renal transplant recipients by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry”,Clin Chem,Oct. 2005, vol. 51,Issue.10,pp. 1903-1904, Oct. 1, 2005, 1903-1904.
Li, Ying et al., “Size Separation of Circulatory DNA in Maternal Plasma Permits Ready Detection of Fetal DNA Polymorphisms”, Clinical Chemistry, 50, 6, 2004, 1002-1011.
Liao, Gary J.W. et al., “Targeted Massively Parallel Sequencing of Maternal Plasma DNA Permits Efficient and Unbiased Detection of Fetal Alleles”, Clinical Chemistry, 57 (1), 2011, 92-101.
Liao, J. et al., “An Alternative Linker-Mediated Polymerase Chain Reaction Method Using a Dideoxynucleotide to Reduce Amplification Background”, Analytical Biochemistry 253, 137-139 (1997).
Lichtenstein, A. V. et al., “Circulating Nucleic Acids and Apoptosis”, Annals New York Academy of Sciences, vol. 945, Aug. 1, 2001, 239-249.
Liew, Michael et al., “Genotyping of Single-Nucleotide Polymorphisms”, Clinical Chemistry, 50(7), 2004, 1156-1164.
Life Technologies, “Ion AmpliSeq Comprehensive Cancer Panel”, 2012, 2 pgs.
Life Technologies, “Ion AmpliSeq™ Designer provides full flexibility to sequence genes of your choice”, 2012, 4 pages.
Liljedahl, Ulrika et al., “Detecting Imbalanced Expression of SNP Alleles by Minisequencing on Microarrays”, BMC Biotechnology, vol. 4, Article No. 24, Oct. 22, 2004, 1-10.
Lindberg, J. et al., “Exome Sequencing of Prostate Cancer Supports the Hypothesis of Independent Tumour Origins”, European Urology, vol. 63, 2013, 347-353.
Lindroos, Katatina et al., “Genotyping SNPs by Minisequencing Primer Extension Using Oligonucleotide Microarrays”, Methods in Molecular Biology, 212, Single Nucleotide Polymorphisms: Methods and Protocols, P-K Kwok (ed.), Humana Press, Inc., Totowa, NJ, 2003, 149-165.
Lo, et al., “Digital PCR for the Molecular Detection of Fetal Chromosomal Aneuploidy”, PNAS, vol. 104, No. 32, Aug. 7, 2007, 13116-13121.
Lo, et al., “Fetal Nucleic Acids in Maternal Blood: the Promises”, Clin. Chem. Lab. Med., 50(6), 2012, 995-998.
Lo, et al., “Free Fetal DNA in Maternal Circulation”, JAMA, 292(23), (Letters to the Editor), 2004, 2835-2836.
Lo, et al., “Next-generation Sequencing of Plasma/Serum DNA: An Emerging Research and Molecular Diagnostic Tool”, Clinical Chemistry, vol. 55, No. 4, Apr. 1, 2009, 607-608.
Lo, “Non-Invasive Prenatal Diagnosis by Massively parallel Sequencing of Maternal Plasma DNA”, Open Biol 2: 120086, 2012, 1-5.
Lo, et al., “Prenatal Sex Determination by DNA Amplification from Maternal Peripheral Blood”, The Lancet,2, 8676, 1989, 1363-1365.
Lo, et al., “Presence of Donor-specific Dna in Plasma of Kidney and Liver-transplant Recipients”, Lancet, vol. 351, No. 9112, May 2, 1998, 1329-1330.
Lo, et al., “Rapid Clearance of Fetal DNA from Maternal Plasma”, Am. J. Hum. Genet., 64, 1999, 218-224.
Lo, et al., “Strategies for the Detection of Autosomal Fetal DNA Sequence from Maternal Peripheral Blood”, Annals New York Academy of Sciences,731, 1994, 204-213.
Lo, et al., “Two-way cell traffic between mother and fetus: biologic and clinical implications”, Blood, 88(11), Dec. 1, 1996, 4390-4395.
Lo, Y M. et al., “Circulating Nucleic Acids in Plasma and Serum: An Overview”, Annals of the New York Academy of Sciences, vol. 945, Sep. 1, 2001, 1-7.
Lo, Y., “Noninvasive prenatal detection of fetal chromosomal aneuploidies by maternal plasma nucleic acid analysis: a review of the current state of the art”, BJOG an International Journal of Obstetrics and Gynaecology, vol. 116, 2009, 152-157.
Lo, Y.M. Dennis, “Fetal Nucleic Acids in Maternal Plasma: Toward the Development of Noninvasive Prenatal Diagnosis of Fetal Chromosomal Aneuploidies”, Ann. N.Y. Acad. Sci., 1137, 2008, 140-143.
Lo, Y.M. Dennis et al., “Maternal Plasma DNA Sequencing Reveals the Genome-Wide Genetic and Mutational Profile of the Fetus”, Science Translational Medicine,, 2 (61), 2010, 13.
Lo, Y.M. Dennis et al., “Plasma placental RNA allelic ratio permits noninvasive prenatal chromosomal aneuploidy detection”, Nature Medicine, 13 (2), 2007, 218-223.
Lo, Y.M. Dennis et al., “Presence of Fetal DNA in Maternal Plasma and Serum”, The Lancet, 350, 1997, 485-487.
Lo, Y.M. Dennis et al., “Quantitative Analysis of Fetal DNA in Maternal Plasma and Serum: Implications for Noninvasive Prenatal Diagnosis”, Am. J. Hum. Genet, 62, 1998, 768-775.
Lo, Y.M.D., “Fetal DNA in Maternal Plasma: Biology and Diagnostic Applications”, Clinical Chemistry, vol. 46, No. 12, 2000, 1903-1906.
Lo, Y.M.D. et al., “Prenatal diagnosis: progress through plasma nucleic acids”, Nature Reviews, vol. 8, 2007, 71-77.
Lo, Y-M D., “Non-invasive prenatal diagnosis using fetal cells in maternal blood”, J. Clin. Pathol., vol. 47, 1994, 1060-1065.
Lo, Y-M.D et al., “Detection of Single-Copy Fetal DNA Sequence from Maternal Blood”, The Lancet, 335, 1990, 1463-1464.
Lo, Y-M.D et al., “Prenatal Determination of Fetal Rhesus D Status by DNA Amplification of Peripheral Blood of Rhesus-Negative Mothers”, Annals New York Academy of Sciences, 731, 1994, 229-236.
Lo, Y-M.D. et al., “Detection of Fetal RhD Sequence from Peripheral Blood of Sensitized RhD-Negative Pregnant Women”, British Journal of Haematology, 87, 1994, 658-660.
Lo, Y-M.D. et al., “Prenatal Determination of Fetal RhD Status by Analysis of Peripheral Blood of Rhesus Negative Mothers”, The Lancet, 341, 1993, 1147-1148.
Loh, Elwyn, “Anchored PCR: Amplification with Single-sided Specificity”, Methods, vol. 2, 1991, pp. 11-19.
Lovmar, L. et al., “Quantitative evaluation by minisequencing and microarrays reveals accurate multiplexed SNP genotyping of whole genome amplified DN”, Nucleic Acids Research, vol. 31, No. 21, 2003, 9 pgs.
Lu, I. et al., “Establishment of a system based on universal multiplex-PCR for screening genetically modified crops”, Anal. Bioanal. Chem, vol. 396, Oct. 24, 2009, 2055-2064.
Lu, S. et al., “Probing Meiotic Recombination and Aneuploidy of Single Sperm Cells by Whole-Genome Sequencing”, Science, vol. 338, Dec. 21, 2012, 1627-1630.
Lui, Y. Y. et al., “Predominant Hematopoietic Origin of Cell-Free DNA in Plasma and Serum after Sex-Mismatched Bone Marrow Transplantation”, Clinical Chemistry, vol. 48, vol. 3, 2002, 421-427.
Lui, Yanni Y. et al., “Circulating DNA in Plasma and Serum: Biology, Preanalytical Issues and Diagnostic Applications”, Clinical Chemistry and Laboratory Medicine, vol. 40, No. 10, Oct. 29, 2002, 962-968.
Lui, Yanni Y. et al., “Origin of Plasma Cell-Free DNA after Solid Organ Transplantation”, Clinical Chemistry, vol. 49, No. 3, Mar. 1, 2003, 495-496.
Lun, Fiona M. et al., “Microfluidics Digital PCR Reveals a Higher Than Expected Fraction of Fetal DNA in Maternal Plasma”, Clinical Chemistry, vol. 54, No. 10, Aug. 14, 2008, 1664-1672.
Lun, Fiona M. et al., “Noninvasive Prenatal Diagnosis of Monogenic Diseases by Digital Size Selection and Relative Mutation Dosage on DNA in Maternal Plasma”, PNAS, 105(50), 2008, 19920-19925.
Ma, Xiaotu et al., “Rise and fall of subclones from diagnosis to relapse in pediatric B-acute lymphoblastic leukaemia”, Nature Communications, vol. 6, Mar. 19, 2015, 1-12.
Mackiewicz, D. et al., “Distribution of Recombination Hotspots in the Human Genome—A Comparison of Computer Simulations with Real Data”, PLOS One, vol. 8, No. 6, Jun. 2013, 11 pages.
Magbanua, M. J. et al., “Abstract PD2-01: Personalized serial circulating tumor DNA (ctDNA) analysis in high-risk early stage breast cancer patients to monitor and predict response to neoadjuvant therapy and outcome in the I-SPY 2 Trial”, Cancer Research, vol. 79, No. 4 Supplement, Feb. 15, 2019.
Mamon, H. et al., “Letters to the Editor: Preferential Amplification of Apoptotic DNA from Plasma: Potential for Enhancing Detection of Minor DNA Alterations in Circulating DNA”, Clinical Chemistry, vol. 54, No. 9, 2008, 1582-1584.
Maniatis, T. et al., “In: Molecular Cloning: A Laboratory Manual”, Cold Spring Harbor Laboratory, New York, Thirteenth Printing, 1986, 458-459.
Mansfield, Elaine S, “Diagnosis of Down Syndrome and Other Aneuploidies Using Quantitative Polymerase Chain Reaction and Small Tandem Repeat Polymorphisms”, Human Molecular Genetics, 2, 1, 1993, 43-50.
Mardis, E. R., “The impact of next-generation sequencing technology on genetics”, Trends in Genetics, vol. 24, No. 3, Feb. 11, 2008, 133-141.
Marguiles, M. et al., “Genome Sequencing in Open Microfabricated High Density Picoliter Reactors”, Nature, vol. 437, No. 7057, Sep. 15, 2005, 376-380.
Margulies, M. et al., “Genome sequencing in microfabricated high-density picolitre reactors”, Nature, vol. 437, Sep. 15, 2005, 376-380.
Margulies, M. et al., “Genome sequencing in microfabricated high-density picolitre reactors plus Supplemental Methods”, Nature, vol. 437, Sep. 15, 2005, 40 pgs.
Marianes, Alexis E. et al., “Targets of Somatic Hypermutation within Immunoglobulin Light Chain Genes in Zebrafish”, Immunology, vol. 132, 2010, pp. 240-255.
Markoulatos, P. et al., “Multiplex Polymerase Chain Reaction: A Practical Approach”, Journal of Clinical Laboratory Analysis, vol. 16, 2002, 47-51.
Maron, Jill L. et al., “Cell-free Fetal DNA Plasma Extraction and Real-time Polymerase Chain Reaction Quantification”, Methods in Molecular Medicine, vol. 132, Aug. 1, 2007, 51-63.
Marshutina, N. V. et al., “Comparative Clinical and Diagnostic Significance of Some Serological Tumor Associated Markers for Different Histological Types of Lung Cancer”, Russian Oncological Journal, vol. 3, 2010, 13-16.
Martinez- Lopez, J. et al., “Real-time PCR Quantification of Haematopoietic Chimerism after Transplantation: A Comparison Between TaqMan and Hybridization Probes Technologies”, International Journal of Laboratory Hematology, vol. 32, Issue 1, Part 1, May 12, 2009, e17-e25.
Martins, et al., “Quantification of Donor-derived DNA in Serum: A New Approach of Acute Rejection Diagnosis in a Rat Kidney Transplantation Model”, Transplantation Proceedings, vol. 37, No. 1,, Jan. 1, 2005, 87-88.
Masuzaki, H. et al., “Detection of cell free placental DNA in maternal plasma: direct evidence from three cases of confined placental mosaicism”, J Med Genet, vol. 41, 2004, 289-292.
Matsubara, T. et al., “Pantropic Retroviral Vectors Integrate and Express in Cells of the Malaria Mosquito, Anopheles gambiae”, PNAS, vol. 93, 1996, pp. 6181-6185.
Matsuzaki, H. et al., “Genotyping over 100,000 SNPs on a pair of oligonucleotide arrays”, Nature Methods, vol. 1, No. 2, Nov. 2004, 109-111.
May, Robert M., “How Many Species Are There on Earth?”, Science, 241, Sep. 16, 1988, 1441-1449.
McBride, D. et al., “Use of Cancer-Specific Genomic Rearrangements to Quantity Disease Burden in Plasma from Patients with Solid Tumors”, Genes, Chromosomes & Cancer, vol. 49, Aug. 19, 2010, 1062-1069.
McCloskey, M. L. et al., “Encoding PCR Products with Batch-stamps and Barcodes”, Biochem Genet., vol. 45, Oct. 23, 2007, 761-767.
McCray, Alexa T. et al., “Aggregating UMLS Semantic Types for Reducing Conceptual Complexity”, MEDINFO 2001: Proceedings of the 10th World Congress on Medical Informatics (Studies in Health Technology and Informatics, 84, V. Patel et al. (eds), IOS Press Amsterdam, 2001, 216-220.
McDonald, B. R. et al., “Abstract P4-01-21: Multiplexed targeted digital sequencing of circulating tumor DNA to detect minimal residual disease in early and locally advanced breast cancer”, Cancer Research, vol. 79, No. 4 Supplement, Feb. 15, 2019.
McDonald, J. P. et al., “Novel thermostable Y-family polymerases: applications for the PCR amplification of damaged or ancient DNAs”, Nucleic Acids Research, vol. 34, No. 4, 2006, 1102-1111.
Mennuti, M. et al., “Is It Time to Sound an Alarm About False-Positive Cell-Free DNA Testing for Fetal Aneuploidy?”, American Journal of Obstetrics, 2013, 5 pgs.
Merriam-Webster, “Medical Definition of Stimulant”, http://www.merriam-webster.com/medical/stimulant, Mar. 14, 2016, 7 pages.
Merriam-Webster, “Universal Definition”, Merriam-Webster.com, 2014, 3 pages.
Mersy, et al., “Noninvasive Detection of Fetal Trisomy 21: Systematic Review and Report of Quality and Outcomes of Diagnostic Accuracy Studies Performed Between 1997 and 2012”, Human Reproduction Update, 19(4), 2013, 318-329.
Mertes, F. et al., “Targeted enrichment of genomic DNA regions for next-generation sequencing”, Briefings in Functional Genomics, vol. 10, No. 6, Nov. 26, 2011, 374-386.
Messmer, Trudy O. et al., “Application of a Nested, Multiplex PCR to Psittacosis Outbreaks”, Journal of Clinical Microbiology, vol. 35, No. 8, 1997, pp. 2043-2046.
Metzker, M. L. et al., “Polymerase Chain Reaction”, Encyclopedia of Medical Devices and Instrumentation, vol. 5, Second Edition, 2006, 380-387.
Metzker, M. L. et al., “Quantitation of Mixed-Base Populations of HIV-1 Variants by Automated DNA Sequencing with BODIPY* Dye-Labeled Primers”, BioTechniques, vol. 25, Sep. 1998, 446-462.
Meuzelaar, Linda S. et al., “Megaplex PCR: A Strategy for Multiplex Amplification”, Nature Methods, vol. 4, 2007, pp. 835-837.
Meyer, M et al., “Illumina Sequencing Library Preparation for Highly Multiplexed Target Capture and Sequencing”, Cold Spring Harbor Protocols, vol. 2010, Issue 6, Jun. 2010, 1-10.
Meyerson, M. et al., “Advances in understanding cancer genomes through second-generation sequencing”, Nature Reviews: Genetics, vol. 11, Oct. 2010, 685-696.
Mikkelsen, T. S. et al., “Genome-wide maps of chromatin state in pluripotent and lineage-committed cells”, Nature, vol. 448, No. 2, Aug. 2007, 553-562.
Milani, et al., “Genotyping Single Nucleotide Polymorphisms by Multiplex Minisequencing Using Tag-arrays”, DNA Microarrays for Biomedical Research, vol. 529, Jan. 16, 2009, 215-229.
Miller, Robert, “Hyperglycemia-Induced Changes in Hepatic Membrane Fatty Acid Composition Correlate with Increased Caspase-3 Activities and Reduced Chick Embryo Viability”, Comparative Biochemistry and Physiology, Part B, 141, 2005, 323-330.
Miller, Robert R., “Homocysteine-Induced Changes in Brain Membrane Composition Correlate with Increased Brain Caspase-3 Activities and Reduced Chick Embryo Viability”, Comparative Biochemistry and Physiology Part B, 136, 2003, 521-532.
Miner, B. E. et al., “Molecular barcodes detect redundancy and contamination in hairpin-bisulfite PCR”, Nucleic Acids Research, vol. 32, No. 17, Sep. 2004, 1-4.
Minkoff, E. et al., “Stem Cells, Cell Division, and Cancer”, Biology Today Third Edition, Chapter 12, 2004, 10 pages.
Miramontes, Pedro et al., “DNA Dimer Correlations Reflect in Vivo Conditions and Discriminate Among Nearest-neighbor Base Pair Free Energy Parameter Measures”, Physica A, vol. 321, 2003, pp. 577-586.
Mitra, S. et al., “Chapter 4 Classification Techniques”, Introduction to Machine Learning and Bioinformatics, First Edition, 2008, 101-127.
Morand, et al., “Hesperidin contributes to the vascular protective effects of orange juice: a randomized crossover study in healthy volunteers”, Am J Clin Nutr. Jan. 2011;93(1 ):73-80. Epub Nov. 10, 2010.
Moreau, Valerie et al., “Zip Nucleic Acids: New High Affinity Oligonucleotides as Potent Primers for PCR and Reverse Transcription”, Nucleic Acids Research, vol. 37, No. 19, e130, 2009, 14 pages.
Moreira, et al., “Increase in and Clearance of Cell-free Plasma DNA in Hemodialysis Quantified by Real-time PCR”, Clinical Chemistry and Laboratory Medicine, vol. 44, No. 12, Dec. 13, 2006, 1410-1415.
Morris, J. K. et al., “Trends in Down's syndrome live births and antenatal diagnoses in England and Wales from 1989 to 2008: analysis of data from the National Down Syndrome Cytogenetic Register”, BMJ Online, vol. 339, Oct. 2009, 5 pages.
Munne, S. et al., “Chromosome Abnormalities in Human Embryos”, Textbook of Assisted Reproductive Techniques, 2004, pp. 355-377.
Munne, S. et al., “Chromosome abnormalities in human embryos”, European Society of Human Reproduction and Embryology: Human Reproduction Update, vol. 4, No. 6, 1998, 842-855.
Munne, S. et al., “Improved implantation after preimplantation genetic diagnosis of aneuploidy”, Reproductive BioMedicine Online, vol 7., No 1., May 15, 2003, 91-97.
Murali, R. et al., “Crystal structure of Taq DNA polymerase in complex with an inhibitory Fab: The Fab is directed against an intermediate in the helix-coil dynamics of the enzyme”, Proc. Natl. Acad. Sci. USA, vol. 95, Oct. 1998, 12562-12567.
Murtaza, M. et al., “Non-Invasive Analysis of Acquired Resistance to Cancer Therapy by Sequencing of Plasma DNA”, Nature (doi:10.1038/nature12065), 2013, 6 pgs.
Muse, Spencer V., “Examining rates and patterns of nucleotide substitution in plants”, Plant Molecular Biology 42: 25-43, 2000.
Myers, Chad L. et al., “Accurate Detection of Aneuploidies in Array CGH and Gene Expression Microarray Data”, Bioinformatics, 20(18), 2004, 3533-3543.
Nagalla, S. R. et al., “Proteomic Analysis of Maternal Serum in Down Syndrome: Identification of Novel Protein Biomarkers”, Journal of Proteome Research, vol. 6, Mar. 21, 2007, 1245-1257.
Namlos, H. M. et al., “Noninvasive Detection of ctDNA Reveals Intratumor Heterogeneity and Is Associated with Tumor Burden in Gastrointestinal Stromal Tumor”, Molecular Cancer Therapeutics, vol. 17, No. 11, 2018, 2473-2480.
Nannya, Yasuhito et al., “A Robust Algorithm for Copy Number Detection Using High-density Oligonucleotide Single Nucleotide Polymorphism Genotyping Arrays”, Cancer Res., 65, 14, 2005, 6071-6079.
Narayan, A. et al., “Ultrasensitive measurement of hotspot mutations in tumor DNA in blood using error-suppressed multiplexed deep sequencing”, Cancer Research, vol. 72, No. 14, Jul. 15, 2012, 3492-3498.
Natera, Inc., “Declaration of Sandra L. Haberny”, May 16, 2019, 3 pages.
Natera, Inc., “Defendant Natera, Inc.'s Invalidity Contentions Under Patent L.R. 3-3; Document Production Accompanying Invalidity Contentions Under Patent L.R. 3-4”, Aug. 20, 2018, 17 pages.
Natera, Inc., “Exhibit 8 Ehrich Invalidity Chart”, Aug. 20, 2018, 16 pages.
Natera, Inc., “Exhibits A-H to Haberny Declaration”, May 16, 2019, 192 pages.
Natera, Inc., “Motion to Dismiss”, May 16, 2019, 2 pages.
Natera, Inc., “Natera Inc.'s First Amended Answer, Affirmative Defenses and Counterclaims”, Aug. 16, 2018, 28 pages.
Natera, Inc., “Natera, Inc.'s Supplemental Objections and Response to Plaintiff Illumina, Inc.'s Interrogatory No. 8”, Mar. 20, 2019, 29 pages.
Natera, Inc., “Opening Brief in Support of Motion to Dismiss”, May 16, 2019, 26 pages.
Natera, Inc., “Petitioner Reply Per Board Order of Nov. 2, 2018 (Paper No. 10)”, Nov. 9, 2018, 8 pgs.
National Institutes of Health, “Genetics Home Reference: Your Guide to Understanding Genetic Conditions”, Feb. 28, 2014, 2 pgs.
Nawroz, H et al., “Microsatellite Alterations in Serum DNA of Head and Neck Cancer Patients”, Nature Medicine, vol. 2, No. 9, Sep. 1996, 1035-1037.
NCBI, “Blast of AAAAAAAAATTTAAAAAAAAATTT”, http://blast.ncbi.nlm.nih.gov/Blast.cgi, 2015, 9 pages.
NCBI, “db SNP rs2056688”, http://www.ncbi.nlm.nih.gov/projects/SNP/snp_ref.cgi?rs=2056688, 2015, 3 pages.
NCBI, “dbSNP record for rs1294331”, Retrieved from the Internet: <URL: www.ncbi.nlm.nih.gov/snp/?term=rs 1294331 >, 2019, 2 pgs.
NCBI, “dbSNP record for rs1872575”, Retrieved from the Internet: <URL: www.ncbi.nlm.nih.gov/snp/?term=rs1872575, 2019, 2 pgs.
NCBI, “dbSNP record for rs2362450”, Retrieved from the Internet: <URL: www.ncbi.nlm.nih.gov/snp/?term=rs2362450>, 2019, 1 pg.
NCBI, “dbSNP record for r52384571”, Retrieved from the Internet: <URL: www.ncbi.nlm.nih.gov/snp/?term=rs2384571>, 2019, 2 pgs.
NCBI, “dbSNP record for rs2498982”, Retrieved from the Internet: <URL: www.ncbi.nlm.nih.gov/snp/?term=rs2498982>, 2019, 3 pgs.
NCBI, “dbSNP record for r53731877”, Retrieved from the Internet: <URL: www.ncbi.nlm.nih.gov/snp/?term=rs3731877>, 2019, 2 pgs.
Neve, B. et al., “Rapid SNP Allele Frequency Determination in Genomic DNA Pools by Pyrosequencing”, BioTechniques, vol. 32, No. 5, May 1, 2002, 1138-1142.
New England Biolabs, “NucleicAcids, Linkers and Primers: Random Primers”, 1998/99Catalog, 1998, 121 and 284.
Newman, A. M. et al., “Integrated digital error suppression for improved detection of circulating tumor DNA”, Nature Biotechnology, vol. 34, No. 5, May 2016, 547-555.
Ng, et al., “Multiplex Sequencing of Paired-end Ditags (MS-PET): A Strategy for the Ultra-high-throughput Analysis of Transcriptomes and Genomes”, Nucleic Acids Research, vol. 34, No. 12, Jul. 13, 2006, 1-10.
Ng, S. B. et al., “Individualised multiplexed circulating tumour DNA assays for monitoring of tumour presence in patients after colorectal cancer surgery”, Scientific Reports, vol. 7, No. 40737, Jan. 19, 2017, 11 pages.
Nguyen-Dumont, T., “A high-plex PCR approach for massively parallel sequencing”, BioTechniques, vol. 55, No. 2, Aug. 2013, 69-74.
Nicolaldes, K. et al., “Noninvasive Prenatal Testing for Fetal Trisomies in a Routinely Screened First-Trimester Population”, American Journal of Obstetrics (article in press), 207, 2012, 1.e1-1.e6.
Nicolaldes, K.H et al., “Validation of Targeted Sequencing of Single-Nucleotide Polymorphisms for Non-Invasive Prenatal Detection of Aneuploidy of Chromosomes 13, 18, 21, X, and Y”, Prenatal Diagnosis, 33, 2013, 575-579.
Nicolaides, Kypros H. et al., “Prenatal Detection of Fetal Triploidy from Cell-Free DNA Testing in Maternal Blood”, Fetal Diagnosis and Therapy, 2013, 1-6.
Nilsson, M. et al., “Padlock Probes: Circularizing Oligonucleotides for Localized DNA Detection”, Science, vol. 265, Sep. 10, 1994, 2085-2088.
Nishigaki, K. et al., “Random PCR-Based Genome Sequencing: A Non-Divide-and-Conquer Strategy”, DNA Research, vol. 7, 2000, 19-26.
Nishiwaki, Morie et al., “Genotyping of Human Papillomaviruses by a Novel One-step Typing Method With Multiplex PCR and Clinical Applications”, Journal of Clinical Microbiology, vol. 46, 2008, pp. 1161-1168.
Nygren, et al., “Quantification of Fetal DNA by Use of Methylation-Based DNA Discrimination”, Clinical Chemistry 56:10 1627-1635 (2010).
O'Connell, G. C. et al., “High Interspecimen Variability in Nucleic Acid Extraction Efficiency Necessitates the Use of Spike-In Control for Accurate qPCR-based Measurement of Plasma Cell-Free DNA Levels”, Lab Medicine, vol. 48, 2017, 332-338.
Oeth, et al., “iPLEX™ Assay: Increased Plexing Efficiency and Flexibility for MassARRAY System Through Single Base Primer Extension with Mass-Modified Terminators”, Sequenom Application Note Doc. No. 8876-006, Apr. 28, 2005, 1-12.
Ogino, S. et al., “Bayesian Analysis and Risk Assessment in Genetic Counseling and Testing”, Journal of Molecular Diagnostics, 6 (1), 2004, 9 pgs.
OHara, O et al., “One-sided Polymerase Chain Reaction: The Amplification of cDNA”, Proceedings of the National Academy of Sciences, vol. 86, 1989, 5673-5677.
Ohira, T. et al., “Tumor volume determines the feasibility of cell-free DNA sequencing for mutation detection in non-small cell lung cancer”, Cancer Science, vol. 107, 2016, 1660-1666.
Ohsawa, M. et al., “Prenatal Diagnosis of Two Pedigrees of Fukuyama Type Congenital Muscular Dystrophy by Polymorphism Analysis”, The Health and Welfare Ministry, 1994, 5 pgs.
Okou, et al., “Microarray-based Genomic Selection for High-throughput Resequencing”, Nature Methods, vol. 4, No. 11, Oct. 14, 2007, 907-909.
Okou, David T. et al., “Combining Microarray-based Genomic Selection (MGS) with the Illumina Genome Analyzer Platform to Sequence Diploid Target Regions”, Annals of Human Genetics, vol. 73, No.5, Aug. 6, 2009, 502-513.
Oliphant, A. et al., “Bead.Array™ Technology: Enabling an Accurate, Cost-Effective Approach to High-Throughput Genotyping”, Bio Techniques, vol. 32, Jun. 2002, S56-S6.
Olivarius, S et al., “High-throughput Verification of Transcriptional starting Sites by Deep-RACE”, Bio Techniques, vol. 46, No. 2, Feb. 2009, 130-132.
Olive, M. et al., “Characterization of the DiFi Rectal Carcinoma Cell Line Derived from a Familial Adenomatous Polyposis Patient”, In Vitro Cellular & Developmental Biology, vol. 29A, No. 3, Part 1, Mar. 1993, 239-248.
Oliver, Dwight H. et al., “Use of Single Nucleotide Polymorphisms (SNP) and Real-time Polymerase Chain Reaction for Bone Marrow Engraftment Analysis”, The Journal of Molecular Diagnostics, vol. 2, No. 4, Nov. 1, 2000, 202-208.
Olivier, et al., “The Invader Assay for SNP Genotyping”, Mutation Research, vol. 573, No. 1-2, Jun. 3, 2005, 103-110.
Olney, R. S. et al., “Chorionic Villus Sampling and Amniocentesis: Recommendations for Prenatal Counseling”, MMWR: Recommendations and Reports, 44(RR-9), Jul. 21, 1995, 1-12.
O'Malley, R. et al., “An adapter ligation-mediated PCR method for high-throughput mapping of T-DNA inserts in the Arabidopsis genome”, Nat. Protoc., 2, 2007, 2910-2917.
Orozco A.F., et al., “Placental Release of Distinct DNA-Associated Micro-Particles into Maternal Circulation: Reflective of Gestation Time and Preeclampsia”, Placenta,30, 2009, 891-897.
Orsouw, et al., “Complexity Reduction of Polymorphic Sequences (Crops): A Novel Approach for Large-scale Polymorphism Discovery in Complex Genomes”, PLoS ONE, vol. 11:e1172, Nov. 14, 2017, 1-10.
Owczarzy, Richard et al., “Melting Temperatures of Nucleic Acids: Discrepancies in Analysis”, Biophysical Chemistry, vol. 117, 2005, pp. 207-215.
Ozawa, Makiko et al., “Two Families with Fukuyama Congenital Muscular Dystrophy that Underwent in Utero Diagnosis Based on Polymorphism Analysis”, Clinical Muscular Dystrophy: Research in Immunology and Genetic Counseling—FY 1994 Research Report, (including copy of text in Japanese), 1994, 8.
Paez, Guillermo J. et al., “Genome coverage and sequence fidelity of Φ29 polymerase-based multiple strand displacement whole genome amplification”, Nucleic Acids Research, 32(9), 2004, 1-11.
Page, S. L. et al., “Chromosome Choreography: The Meiotic Ballet”, Science, 301, 2003, 785-789.
Paik, P. K. et al., “Next-Generation Sequencing of Stage IV Squamous Cell Lung Cancers Reveals an Association of P13K Aberrations and Evidence of Clonal Heterogeneity in Patients with Brain Metastases”, Cancer Discovery, vol. 5, Apr. 30, 2015, 610-621.
Pakstis, et al., “Candidate SNPs for a Universal Individual Identification Panel”, Human Genetics, vol. 121, No. 3-4,, Feb. 27, 2007, 305-317.
Pakstis, et al., “SNPS for Individual Identification”, Forensic Science International, vol. 1, May 22, 2008, 479-481.
Palka-Santini, Maria et al., “Large Scale Multiplex PCR Improves Pathogen Detection by DNA Microarrays”, BMC Microbiology, vol. 9, No. 1, 2009, 14 pages.
Palomaki, G. E. et al., “DNA sequencing of maternal plasma to detect Down syndrome: An international clinical validation study”, Genetics in Medicine, vol. 13, No. 1, Nov. 2011, 913-920.
Palomaki, Glenn et al., “DNA Sequencing of Maternal Plasma Reliably Identifies Trisomy 18 and Trisomy 13 as Well as Down Syndrome: an International Collaborative Study”, Genetics in Medicine, 2012, 10.
Palomaki, Glenn E. et al., “DNA Sequencing of Maternal Plasma to Detect Down Syndrome: An International Clinical Validation Study”, Genetics in Medicine (pre-print version), 13, 2011, 8 pgs.
Panjkovich, Alejandro et al., “Comparison of Different Melting Temperature Calculation Methods for Short DNA Sequences”, Bioinformatics, vol. 21, 2005, pp. 711-722.
Papadopoulou, E. et al., “Cell-Free DNA and RNA in Plasma as a New Molecular Marker for Prostate Cancer”, Oncology Research, vol. 14, 2004, 439-445.
Papageorgiou, Elisavet A. et al., “Fetal-Specific DNA Methylation Ratio Permits Noninvasive Prenatal Diagnosis of Trisomy 21”, Nature Medicine (advance online publication),17, 2011, 5 pgs.
Parameswaran, P. et al., “A pyrosequencing-tailored nucleotide barcode design unveils opportunities for large-scale sample multiplexing”, Nucleic Acids Research, vol. 35, No. 19, Oct. 11, 2007, 9 pages.
Park, et al., “First-Line Erlotinib Therapy Until and Beyond Response Evaluation Criteria in Solid Tumors Progression in Asian Patients With Epidermal Growth Factor Receptor Mutation-Positive Non-Small-Cell Lung Cancer”, JAMA Oncol., 2(3), 2015, 305-312.
Parker, A. V. et al., “The Effect of Sodium Citrate on the Stimulation of Polymorphonuclear Leukocytes”, Investigative Ophthalmology & Visual Science, vol. 26, 1985, 1257-1261.
Paruzynski, A. et al., “Genome-wide high-throughput integrome analyses by nrLAM-PCR and next-generation sequencing”, Nature Protocols, vol. 5, No. 8, Jul. 8, 2010, 1379-1395.
Pask, R. et al., “Investigating the utility of combining 29 whole genome amplification and highly multiplexed single nucleotide polymorphism BeadArray TM genotyping”, BMC Biotechnology, vol. 4, No. 15, Jul. 27, 2004, 8 pages.
Pastinen, T. et al., “Minisequencing: A Specific Tool for DNA Analysis and Diagnostics on Oligonucleotide Arrays”, Genome Research, vol. 7, 1997, 606-614.
Pathak, A. et al., “Circulating Cell-Free DNA in Plasma/Serum of Lung Cancer Patients as a Potential Screening and Prognostic Tool”, Clinical Chemistry, 52, 2006, 1833-1842.
Patil, N. et al., “Blocks of Limited Haplotype Diversity Revealed by High-Resolution Scanning of Human Chromosome 21”, Science, vol. 294, Nov. 23, 2001, 1719-1723.
Paunio, T. et al., “Preimplantation diagnosis by whole-genome amplification, PCR amplification, and solid-phase minisequencing of blastomere DNA”, Clinical Chemistry, vol. 42, No. 9, 1996, 1382-1390.
PCT/US2006/045281, “International Preliminary Report on Patentability”, mailed May 27, 2008, 1 pg.
PCT/US2006/045281, “International Search Report and Written Opinion”, mailed Sep. 28, 2007, 7 pgs.
PCT/US2008/003547, “International Search Report”, mailed Apr. 15, 2009, 5 pgs.
PCT/US2009/034506, “International Search Report”, mailed Jul. 8, 2009, 2 pgs.
PCT/US2009/045335, “International Search Report”, mailed Jul. 27, 2009, 1 pg.
PCT/US2009/052730, “International Search Report”, mailed Sep. 28, 2009, 1 pg.
PCT/US2010/050824, “International Search Report”, mailed Nov. 15, 2010, 2 pgs.
PCT/US2011/037018, “International Search Report”, mailed Sep. 27, 2011, 2 pgs.
PCT/US2011/061506, “International Search Report”, mailed Mar. 16, 2012, 1 pgs.
PCT/US2011/066938, “International Search Report”, mailed Jun. 20, 2012, 1 pg.
PCT/US2012066339, “International Search Report”, mailed Mar. 5, 2013, 1 pg.
PCT/US2013/028378, “International Search Report and Written Opinion”, mailed May 28, 2013, 11 pgs.
PCT/US2013/57924, “International Search Report and Written Opinion”, mailed Feb. 18, 2014, 8 pgs.
PCT/US2014/051926, “International Search Report and Written Opinion”, Dec. 9, 2014, 3 pgs.
Pearson, K., “On the criterion that a given system of deviations from the probable in the case of a correlated system of variables is such that it can be reasonably supposed to have arisen from random sampling”, Philosophical Magazine Series 5, vol. 50, Issue 302, 1900, 157-175.
Pelizzari, C. A. et al., “Quantitative analysis of DNA array autoradiographs”, Nucleic Acids Research, vol. 28, No. 22, 2000, 4577-4581.
Pena, Sergio D.J et al., “Paternity Testing in the DNA Era”, Trends in Genetics, 10, 6, 1994, 204-209.
Perakis, S. et al., “Advances in Circulating Tumor DNA Analysis”, Advances in Clinical Chemistry, vol. 80, 2017, 73-153.
Pergament, E. et al., “Single-Nucleotide Polymorphism-Based Noninvasive Prenatal Screening in a High-Risk and Low-Risk Cohort”, Obstetrics & Gynecology, vol. 124, No. 2, Part 1, Aug. 2014, 210-218 + Appendices.
Perkel, Jeffrey M., “Overcoming the Challenges of Multiplex PCR”, Biocompare Editorial Article, 2012, 1-5.
Perry, George H. et al., “The Fine-Scale and Complex Architecture of Human Copy-Number Variation”, The American Journal of Human Genetics,82, 2008, 685-695.
Pertl, B. et al., “Detection of Male and Female Fetal DNA in Maternal Plasma by Multiplex Fluorescent Polymerase Chain Reaction Amplification of Short Tandem Repeats”, Hum. Genet, 106, 2000, 4549.
Peters, D., “List of Materials Considered by David Peters, Ph.D.”, Jun. 13, 2019, 2 pages.
Peters, David P. et al., “Noninvasive Prenatal Diagnosis of a Fetal Vlicrodeletion Syndrome”, New England Journal of Medicine, 365(19), 2011, 1847-1848.
Pfaffl, Michael W., “Quantification Strategies in Real-time PCR”, A-Z of quantitative PCR, 2004, pp. 87-112.
Pfaffl, Michael W., “Relative Expression Software Tool (REST ©) for Group-Wise Comparison and Statistical Analysis of Relative Expression Results in real-Time PCR”, Nucleic Acids Research, 30(9), 2002, 10 pgs.
Philip, J. et al., “Late First-Trimester Invasive Prenatal Diagnosis: tesults of an International Randomized Trial”, American College of Obstetricians and Gynecologists, vol. 103, No. 6, Jun. 2004, 1164-1173.
Phillips, C. et al., “Resolving Relationship Tests that Show Ambiguous STR Results Using Autosomal SNPs as Supplementary Markers”, Forensic Science International: Genetics 2, 2008, 198-204.
Pinard, et al., “Assessment of Whole Genome Amplification-induced Bias Through High-throughput, Massively Parallel Whole Genome Sequencing”, BMC Genomics, vol. 72216, Aug. 23, 2006, 1-21.
Pirker, C. et al., “Whole Genome Amplification for CGH Analysis: Linker-Adapter PCR as the Method of Choice for Difficult and Limited Samples”, Cytometry Part A, vol. 61 A, 2004, 26-34.
Podder, Mohua et al., “Robust SN P genotyping by multiplex PCR and arrayed primer”, BMC Medical Genomics,1(5), 2008, 1-15.
Poirier, K. et al., “Maternal mosaicism for mutations in the ARX gene in a family with X linked mental retardation”, Human Genetics, vol. 118, Aug. 3, 2005, 45-48.
Pont-Kingdon, G. et al., “Rapid Detection of Aneuploidy (Trisomy 21) by Allele Quantification Combined with Melting Curves Analysis of Single-Nucleotide Polymorphism Loci”, Clinical Chemistry, vol. 49, No. 7, 2003, 1087-1094.
Poon, L. L. et al., “Differential DNA Methylation between Fetus and Mother as a Strategy for Detecting Fetal DNA in Maternal Plasma”, Clinical Chemistry, vol. 48, No. 1, 2002, 35-41.
Popova, T. et al., “Genome Alteration Print (GAP): a tool to visualize and mine complex cancer genomic profiles obtained by SNP arrays”, Genome Biology, vol. 10, R128, Nov. 11, 2009, 1-14.
Porreca, Gregory J et al., “Multiplex Amplification of Large Sets of Human Exons”, Nature Methods, 4, (advance online publication), 2007, 6.
Pourmand, et al., “Multiplex Pyrosequencing”, Nucleic Acid Research, vol. 30, No. 7, Apr. 1, 2002, 1-5. s.
Prabhu, et al., “Overlapping Pools for High-throughput Targeted Resequencing”, Genome Research, vol. 19, May 15, 2009, 1254-1261.
Price, T.S. et al., ““SW-ARRAY: a dynamic programming solution for the identification of copy-number changes in genomic DNA using array comparative genome hybridization data”,”, Nucleic Acids Research, vol. 33, No. 11, Jun. 16, 2005, 3455-3464.
Primdahl, H. et al., “Allelic Imbalances in Human Bladder Cancer: Genome-Wide Detection With High-Density Single-Nucleotide Polymorphism Arrays”, Journal of the National Cancer Institute, vol. 94, No. 3, Feb. 6, 2002, 216-223.
Profitt, J et al., “Isolation and Characterisation of Recombination Events Involving Immunoglobulin Heavy Chain Switch Regions in Multiple Myeloma Using Long Distance Vectorette PCR (Ldv-pcr)”, Leukemia, vol. 13, No. 7, Jul. 1999, 1100-1107.
Puszyk, William M. et al., “Noninvasive Prenatal Diagnosis of Aneuploidy Using Cell-tree Nucleic Acids in Maternal Blood: Promises and Unanswered Questions”, Prenatal Diagnosis, vol. 28, No. 1, Nov. 16, 2007, 1-6.
Qiagen, “QIAamp DNA Mini Kit and QIAamp DNA Blood Mini Kit Handbook”, QIAamp DNA Mini Kit and QIAamp DNA Blood Mini Kit Handbook, Feb. 2003 (“Qiagen (2003)”), 2003, 68 pages.
Qin, Z. S. et al., “Partition-Ligation-Expectation-Maximization Algorithm for Haplotype Inference with Single-Nucleotide Polymorphisms”, Am. J. Hum Genet., vol. 71, 2002, 1242-1247.
Quan, P. C. et al., “Studies on the mechanism of NK cell lysis”, The Journal of Immunology, vol. 128, 1982, 1786-1791.
Quinlan, M. P., “Amniocentesis: Indications and Risks”, American Medical Association Journal of Ethics: Virtual Mentor, vol. 10, No. 5, May 2008, 304-306.
Quinn, G. P. et al., “Experimental Design and Data Analysis for Biologists”, Graphical Exploration of Data, 2002, 64-67.
Rabinowitz, et al., “Accurate Prediction of HIV-1 Drug Response from the Reverse Transcriptase and Protease Amino Acid Sequences Using Sparse Models Created by Convex Optimization”, Bioinformatics, 22, 5, 2006, 541-549.
Rabinowitz, M., “A System and Method for Integrating, Validating and Applying Genetic and Clinical Data to Enhance Medical Decisions”, Nov. 29, 2005, 155 pgs.
Rabinowitz, Matthew et al., “Origins and rates of aneuploidy inhuman blastomeres”, Fertility and Sterility, vol. 97, No. 2, Feb. 2012, 395-401.
Rabinowitz, Matthew. et al., “Non-Invasive Prenatal Aneuploidy Testing of Chromosomes 13, 18, 21, X, and Y Using Targeted Sequencing of Polymorphic Loci”, The American Society of Human Genetics, meeting poster, 2012, 1 pg.
Rachlin, J. et al., “Computational tradeoffs in multiplex PCR assay design for SNP genotyping”, BMC Genomics, vol. 6, No. 102, Jul. 25, 2005, 11 pages.
Ragoussis, J., “Genotyping Technologies for Genetic Research”, Annual Review of Genomics and Human Genetics, vol. 10 (1), Sep. 1, 2009, 117-133.
Rahmann, Sven et al., “Mean and variance of the Gibbs free energy of oligonucleotides in the nearest neighbor model under varying conditions”, Bioinformatics, 20(17), 2004, 2928-2933.
Raindance Technologies, “Multiplexing with RainDrop Digital PCR”, Application Note, 2013, 2 pgs.
Raindance Technologies, et al., “RainDance Technologies Introduces the RDT 1000”, RainDance Technologies, Nov. 12, 2008.
Rava, Richard P. et al., “Circulating Fetal Cell-Free DNA Fraction Differ in Autosomal Aneuploidies and Monosomy X”, Clinical Chemistry, 60(1), (papers in press), 2013, 8 pgs.
Ravipati, Goutham et al., “Comparison of Sensitivity, Specificity, Positive Predictive Value, and Negative Predictive Value of Stress Testing Versus 64-Multislice Coronary Computed Tomography Angiography in Predicting Obstructive Coronary Artery Disease Diagnosed by Coronary Angiogr”, The American Journal of Cardiology, Coronary Artery Disease. vol. 101, Issue 6, Mar. 15, 2008, 774-775.
Rechitsky, Svetiana et al., “Preimplantation Genetic Diagnosis with HLA Matching”, Reproductive Bio Medicine Online, 9, 2, 2004, 210-221.
Reeves, R. H. et al., “Too much of a good thing: mechanisms of gene action in Down syndrome”, Trends in Genetics, vol. 17, No. 2, Feb. 2, 2001, 83-88.
Reinert, T. et al., “Analysis of circulating tumour DNA to monitor disease burden following colorectal cancer surgery”, Gut, vol. 65, 2016, 625-634.
Renwick, P. et al., “Proof of Principle and First Cases Using Preimplantation Genetic Haplotyping—A Paradigm Shift for Embryo Diagnosis”, Reproductive BioMedicine Online, 13 (1), 2006, 110-119.
Rhoads, A. et al., “PacBio Sequencing and Its Applications”, Genomics Proteomics Bioinformatics, vol. 13, Nov. 2, 2015, 278-289.
Ricciotti, Hope, “Eating by Trimester”, Online]. Retrieved from Internet:<http://www.youandyourfamily.com/article.php?story=Eating+by+Trimester>, 2014, 3.
Riley, D. E., “DNA Testing: An Introduction for Non-Scientists an Illustrated Explanation”, Scientific Testimony: An Online Journal, http://www.scientific.org/tutorials/articles/riley/riley.html, Apr. 6, 2005, 22 pages.
Riva, F., “Patient-Specific Circulating Tumor DNA Detection during Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer”, Clinical Chemistry, vol. 63, No. 3, 2017, 691-699.
Robertson, G. et al., “Genome-wide profiles of STAT1 DNA association using chromatin immunoprecipitation and massively parallel sequencing”, Nature Methods, vol. 4, No. 8, Aug. 2007, 651-657.
Roche Diagnostics, et al., “Versatile Nucleic Acid Purification”, MagnaPure Manual, Feb. 3, 2012.
Rogaeva, E. et al., “The Solved and Unsolved Mysteries of the Genetics of Early-Onset Alzheimer's Disease”, NeuroMolecular Medicine, vol. 2, 2002, 1-10.
Roman, B. L. et al., “Non-Radioisotopic AFLP Method Using PCR Primers Fluorescently Labeled with CyA 5”, BioTechniques, vol. 26, Feb. 1999, 236-238.
Roper, Stephen M. et al., “Forensic Aspects of DNA-Based Human Identity Testing”, Journal of Forensic Nursing, 4, 2008, 150-156.
Rosado, J. A. et al., “Tyrosine kinases activate store-mediated Ca2+ entry in human platelets through the reorganization of the actin cytoskeleton”, Biochem. J., vol. 351, 2000, 429-437.
Rosen, D. R. et al., “Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis”, Nature, vol. 362, Mar. 4, 1993, 59-62.
Ross, P. et al., “Quantitative Approach to Single-Nucleotide Polymorphism Analysis Using MALDI-TOF Mass Spectrometry”, BioTechniques, vol. 29, Sep. 2000, 620-629.
Rothberg, et al., “The Development and Impact of 454 Sequencing”, Nature Biotechnology, vol. 26, No. 10, Oct. 9, 2008, 1117-1124.
Roux, K., “Optimization and Troubleshooting in PCR”, PCR Methods Appl. 4, 1995, 185-194.
Rozen, Steve et al., “Primer3 on the WWW for General Users and for Biologis Programmers”, Methods in Molecular Biology, 132: Bioinformatics Methods and Protocols, 1999, 365-386.
Ruano, G. et al., “Haplotype of multiple polymorphisms resolved by enzymatic amplification of single DNA molecules”, Proc. Nati. Acad. Sci. USA, vol. 87, Aug. 1990, 6296-6300.
Rubio, J. M. et al., “Semi-nested, Multiplex Polymerase Chain Reaction for Detection of Human Malaria Parasites and Evidence of Plasmodium Vivax infection in Equatorial Guinea”, The American Journal of Tropical Medicine and Hygiene, vol. 60, 1999, pp. 183-187.
Ruschendorf, et al., “Alohomora: A Tool for Linkage Analysis Using 10K SNP Array Data”, Bioinformatics Applications Notes, vol. 21, No. 9, Jan. 12, 2005, 2123-2125.
Russell, L. M., “X Chromosome Loss and Ageing”, Cytogenetic and Genome Res., 116, 2007, 181-185.
Ryan, A. et al., “Informatics-Based, Highly Accurate, Noninvasive Drenatal Paternity Testing”, Genetics in Medicine (advance online publication), 2012, 5 pgs.
Ryan, B. M. et al., “A prospective study of circulating mutant KRAS2 in he serum of patients with colorectal neoplasia: strong prognostic ndicator in postoperative follow up”, Gut, vol. 52, 2003, 101-108.
Rychlik, et al., “Optimization of the annealing temperature for DNA amplification in vitro”, Nucleic Acids Research, 18(21), 1990, 6409-6412.
Sahota, A., “Evaluation of Seven PCR-Based Assays for the Analysis of Microchimerism”, Clinical Biochemistry, vol. 31, No. 8., 1998, 641-645.
Sahukhal, G. S. et al., “msaABCR operon positively regulates biofilm development by repressing proteases and autolysis in Staphlococcus aureus”, FEMS Microbiology Letters, vol. 362, No. 4, 2015, 1-10.
Saito, H. et al., “Prenatal DNA diagnosis of a single-gene disorder from maternal plasma”, The Lancet, vol. 356, Sep. 30, 2000, 1170.
Saker, A. et al., “Genetic characterisation of circulating fetal cells allows non-invasive prenatal diagnosis of cystic fibrosis”, Prenatal Diagnosis, vol. 26, Jul. 11, 2006, 906-916.
Samango-Sprouse, C. et al., “SNP-Based Non-Invasive Prenatal Testing Detects Sex Chromosome Aneuploidies with High Accuracy”, Prenatal Diagnosis, 33, 2013, 1-7.
Samura, O. et al., “Diagnosis of Trisomy 21 in Fetal Nucleated Erythrocytes from Maternal Blood by Use of Short Tandem Repeat Sequences”, Clinical Chemistry, vol. 47, No. 9, 2001, 1622-1626.
Sanchez, C. et al., “New insights into structural features and optimal detection of circulating tumor DNA determined by single-strand DNA analysis”, Nature Partner Journals, vol. 3, No. 31, Nov. 23, 2018, 12 pgs.
Sander, Chris , “Genetic Medicine and the Future of Health Care”, Science, 287(5460), 2000, 1977-1978.
Sanger, et al., “Nucleotide Sequence of Bacteriophage Lambda DNA”, Journal of Molecular Biology, vol. 162, No. 4, Dec. 25, 1982, 729-773.
Santalucia, J. et al., “The Thermodynamics of DNA Structural Motifs”, Annu. Rev. Biophys. Biomol. Struct., 33, 2004, 415-440.
Santalucia, John J.R et al., “Improved Nearest-Neighbor Parameters for Predicting DNA Duplex Stability”, Biochemistry, 35, 1996, 3555-3562.
Santalucia, JR., J., “Physical Principles and Visual-OMP Software for Optimal PCR Design”, Methods in Molecular Biology, vol. 402, 2007, 3-33.
Sasabe, Yutaka, “Genetic Diagnosis of Gametes and Embryos Resulting from ART”, Japanese Journal of Fertility and Sterility, vol. 46, No. 1, 2001, 43-46.
Scarpa, A. et al., “Molecular Typing of Lung Adenocarcinoma on Cytological Samples Using a Multigene Next Generation Sequencing Panel”, PLOS One, vol. 8, No. 11, Nov. 13, 2013, 6 pgs.
Schaaf, C. P. et al., “Copy Number and SNP Arrays in Clinical Diagnostics”, Annu. Rev. Genomics Hum. Genet, vol. 12, 2011, 25-51.
Scheet, P. et al., “A Fast and Flexible Statistical Model for Large-Scale Population Genotype Data: Applications to Inferring Missing Genotypes and Haplotypic Phase”, The American Journal of Human Genetics, vol. 78, Apr. 2006, 629-644.
Schmitt, M. W. et al., “Detection of ultra-rare mutations by next-generation sequencing”, PNAS, vol. 109, No. 36, Sep. 4, 2012, 14508-14513.
Schoske, R et al., “Multiplex PCR Design Strategy used for the Simultaneous Amplification of 10 Y Chromosome Short Tandem Repeat (STR) Loci”, Analytical and Bioanalytical Chemistry, vol. 375, 2003, 333-343.
Schoumans, J et al., “Detection of chromosomal imbalances in children with idiopathic mental retardation by array based comparative genomic hybridisation (array-CGH)”, JMed Genet, 42, 2005, 699-705.
Schubert, “Picking out prenatal DNA”, Nature Medicine, vol. 10, No. 785, Aug. 2004, 1 page.
Schwarzenbach, H. et al., “Cell˜free nucleic acids as biomarkers in cancer patients”, Nature Reviews: Cancer, vol. 11, Jun. 2011, 426-437.
Schwarzenbach, H. et al., “Detection and Characterization of Circulating Microsatellite-DNA in Blood of Patients with Breast Cancer”, Ann. NY. Acad. Sci., vol. 1022, 2004, 25-32.
Schwarzenbach, H. et al., “Evaluation of cell-free tumour DNA and RNA in patients with breast cancer and benign breast disease”, Molecular BioSystems, vol. 7, 2011, 2848-2854.
Sebat, Jonathan et al., “Strong Association of De Novo Copy Number Mutations with Autism”, Science, 316, 2007, 445-449.
Sehnert, A. et al., “Optimal Detection of Fetal Chromosomal Abnormalities by Massively Parallel DNA Sequencing of Cell-Free Fetal DNA from Maternal Blood”, Clinical Chemistry (papers in press), 57 (7), 2011, 8 pgs.
Seppo, A. et al., “Detection of circulating fetal cells utilizing automated microscopy: potential for noninvasive prenatal diagnosis of chromosomal aneuploidies”, Prenatal Diagnosis, vol. 28, Jul. 22, 2008, 815-821.
Sermon, Karen et al., “Preimplantation genetic diagnosis”, The Lancet, Lancet Limited. 363(9421), 2000, 1633-1641.
Servin, B et al., “MOM: A Program to Compute Fully Informative Genotype Frequencies in Complex Breeding Schemes”, Journal of Heredity, vol. 93, No. 3, Jan. 1, 2002 (Jan. 1, 2002), pp. 227-228.
Sham, P. et al., “DNA Pooling: A Tool for Large-Scale Association Studies”, Nature Reviews Genetics, vol. 3, Nov. 2002, 862-871.
Shapero, M. H. et al., “MARA: A Novel Approach for Highly Multiplexed Locus-specific SNP Genotyping Using High-density DNA Oligonucleotide Arrays”, Nucleic Acids Research, vol. 32, No. 22, 2004, 1-9.
Sharples, et al., “Diagnostic Accuracy of Coronary Angiography and Risk Factors for Post-heart-transplant Cardiac Allograft Vasculopathy”, Transplantation, vol. 76, No. 4, Aug. 27, 2003, 679-682.
Shaw-Smith, et al., “Microarray Based Comparative Genomic Hybridisation (array-CGH) Detects Submicroscopic Chromosomal Deletions and Duplications in Patients with Learning Disability/Mental Retardation and Dysmorphic Features”, J. Med. Genet, 41, 2004, 241-248.
Shen, et al., “High-quality DNA sequence capture of 524 disease candidate genes”, High-quality DNA sequence capture of 524 disease candidate genes, Proceedings of the National Academy of Sciences, vol. 108, No. 16, Apr. 5, 2011 (Apr. 5, 2011), pp. 6549-6554.
Shen, R. et al., “High-throughput SNP genotyping on universal bead arrays”, Mutation Research, vol. 573, Feb. 11, 2005, 70-82.
Shen, Zhiyong, “MPprimer: a program for reliable multiplex PCR primer design”, BMC Bioinformatics 2010, 11:143, 1-7.
Shendure, J. et al., “Accurate Multiplex Polony Sequencing of an Evolved Bacterial Genome”, Science, Nov. 30, 2007, 18-24.
Shendure, J. et al., “Next-generation DNA sequencing”, Nature Biotechnology, vol. 26, No. 10, Oct. 2008, 1135-1145.
Sherlock, J et al., “Assessment of Diagnostic Quantitative Fluorescent Multiplex Polymerase Chain Reaction Assays Performed on Single Cells”, Annals of Human Genetics,62, 1, 1998, 9-23.
Shi, H. et al., “Melanoma whole-exome sequencing identifies V600E B-RAF amplification-mediated acquired B-RAF inhibitor resistance”, Nature Communications, vol. 3, No. 724, Mar. 6, 2012, 8 pages.
Shinozaki, M. et al., “Utility of Circulating B-RAF DNA Mutation in Serum for Monitoring Melanoma Patients Receiving Biochemotherapy”, Clin Cancer Res, vol. 13, No. 7, Apr. 1, 2007, 2068-2074.
Shiroguchi, K. et al., “Digital RNA sequencing minimizes sequence-dependent bias and amplification noise with optimized single-molecule barcodes”, PNAS, vol. 109, No. 4, Jan. 24, 2012, 1347-1352.
Shokralla, S. et al., “Next-generation DNA barcoding: using next-generation sequencing to enhance and accelerate DNA barcode capture from single specimens”, Molecular Ecology Resources, vol. 14, 2014, 892-901.
Short, N. J. et al., “Targeted next-generation sequencing of circulating cell-free DNA vs bone marrow in patients with acute myeloid leukemia”, Blood Advances, vol. 4, No. 8, Apr. 23, 2020, 1670-1677.
Shyamala, Venkatakrishna et al., “Genome Walking by Single-Specific-Primer Polymerase Chain Reaction: SSP-PCR”, Gene, vol. 84, 1989, pp. 1-8.
Siebert, P. D. et al., “An improved PCR method for walking in uncloned genomic DNA”, Nucleic Acids Research, vol. 23, No. 6, 1995, 1087-1088.
Sigdel, T. et al., “Plasma Donor-Derived Cell-Free DNA Quantification by massively multiplex PCR Distinguishes Kidney Transplant Acute Rejection”, Transplantation, vol. 102, No. 78, Jul. 2018, 8178-8179.
Sigdel, T. K. et al., “Optimizing Detection of Kidney Transplant Injury by Assessment of Donor-Derived Cell-Free DNA via Massively Multiplex PCR”, Journal of Clinical Medicine, vol. 8, No. 19, Dec. 23, 2018, 17 pages.
Simpson, J. et al., “Fetal Cells in Maternal Blood: Overview and Historical Perspective”, Annals New York Academy of Sciences, 731, 1994, 1-8.
Singh, Vinayak K. et al., “PCR Primer Design”, Molecular Biology Today, vol. 2, 2001, pp. 27-32.
Sint, Daniela et al., “Advances in Multiplex PCR: Balancing Primer Efficiencies and Improving Detection Success”, Methods in Ecology and Evolution, 3, 2012, 898-905.
Sivertsson, A. et al., “Pyrosequencing as an Alternative to Single-Strand Conformation Polymorphism Analysis for Detection of N-ras Mutations in Human Melanoma Metastases”, Clinical Chemistry, vol. 48, No. 12, 2002, 2164-2170.
Slater, Howard et al., “High-Resolution Identification of Chromosomal Abnormalities Using Oligonucleotide Arrays Containing 116,204 SNPs”, Am. J. Hum. Genet., 77, 5, 2005, 709-726.
Smith, et al., “Rapid Whole-genome Mutational Profiling using Next-generation Sequencing Technologies”, Genome Research, vol. 18, Sep. 4, 2008, 1638-1642.
Smith, James F. et al., “Cell-free Fetal DNA in Maternal Plasma”, Neo Reviews, vol. 9, No. 8, Aug. 1, 2008, e332-e337.
Snijders, Antoine et al., “Assembly of Microarrays for Genome-Wide Measurement of DNA Copy Number”, Nature Genetic, 29, 2001, 263-264.
Societies Related to Genetic Med, “Guideline related to genetic examination”, Japanese Society for Genetic Counseling, Japanese Society for Gene Diagnosis and Therapy, Japan Society of Obstetrics and Gynecology, 2003, 2-15.
Solexa, “Application Note: DNA Sequencing”, 2006, 1-2.
Solomon, M. J. et al., “Formaldehyde-mediated DNA-protein crosslinking: A probe for in vivo chromatin structures”, Proc. Natl. Acad. Sci. USA, vol. 82, 1985, 6470-6474.
Sorenson, G. D. et al., “Soluble Normal and Mutated DNA Sequences from Single-Copy Genes in Human Blood”, Cancer Epdemiology, Biomarkers & Prevention, vol. 3, Jan./Feb. 1994, 67-71.
sourceforge.net, “Primer3”, http://primer3.sourceforge.net/, 2009, 1 pg.
Sparks, A. et al., “Non-Invasive Prenatal Detection and Selective Analysis of Cell-Free DNA Obtained from Maternal Blood: Evaluation for Trisomy 21 and Trisomy 18”, American Journal of Obstetrics & Gynecology 206, 2012, 319.e1-319.e9.
Sparks, Andrew B. et al., “Selective Analysis of Cell-Free DNA in Maternal Blood for Evaluation of Fetal Trisomy”, Prenatal Diagnosis, 32, 2012, 1-7.
Spencer, K. et al., “Maternal serum levels of dimeric inhibin A in pregnancies affected by trisomy 21 in the first trimester”, Prenatal Diagnosis, vol. 21, 2001, 441-444.
Spencer, K. et al., “Maternal serum levels of total activin-A in first-trimester trisomy 21 pregnancies”, Prenatal Diagnosis, vol. 21, 2001, 270-273.
Spertini, D. et al., “Screening of Transgenic Plants by Amplification of Unknown Genomic DNA Flanking T-DNA”, BioTechniques, vol. 27, Aug. 1999, 308-314.
Spes, et al., “Diagnostic and Prognostic Value of Serial Dobutamine Stress Echocardiography for Noninvasive Assessment of Cardiac Allograft Vasculopathy: A Comparison With Coronary Angiography and Intravascular Ultrasound”, Circulation, vol. 100, No. 5, Aug. 3, 1999, 509-515.
Spindler, K.L. G. et al., “Cell-free DNA in healthy individuals, noncancerous disease and strong prognostic value in colorectal cancer”, International Journal of Cancer, vol. 135, 2014, 2984-2991.
Spindler, K.L. G. et al., “Cell-Free DNA in Metastatic Colorectal Cancer: A Systematic Review and Meta-Analysis”, The Oncologist, vol. 22, 2017, 1049-1055.
Spiro, Alexander et al., “A Bead-Based Method for Multiplexed Identification and Quantitation of DNA Sequences Using Flow Cytometry”, Applied and Environmental Microbiology, 66, 10, 2000, 4258-4265.
Spits, C et al., “Optimization and Evaluation of Single-Cell Whole Genome Multiple Displacement Amplification”, Human Mutation, 27(5), 496-503, 2006.
Srinivasan, et al., “Noninvasive Detection of Fetal Subchromosome Abnormalities via Deep Sequencing of Maternal Plasma”, The American Journal of Human Genetics 92, 167-176, Feb. 7, 2013.
Stephens, M. et al., “Accounting for Decay of Linkage Disequilibrium in Haplotype Inference and Missing-Data Imputation”, Am. J. Hum. Genet, vol. 76, 2005, 449-462.
Stephens, Mathews. et al., “A Comparison of Bayesian Methods for Haplotype Reconstruction from Population Genotype Data”, Am. J. Hum. Genet.,73, 2003, 1162-1169.
Stevens, Robert et al., “Ontology-Based Knowledge Representation for Bioinformatics”, Briefings in Bioinformatics, 1, 4, 2000, 398-414.
Stewart, C. M. et al., “Circulating cell-free DNA for non-invasive cancer management”, Cancer Genetics, vol. 228-229, 2018, 169-179.
Stewart, S. et al., “Revision of the 1990 Working Formulation for the Standardization of Nomenclature in the Diagnosis of Heart Rejection”, The Journal of Heart and Lung Transplantation, vol. 24, No. 11, Nov. 2005, 1710-1720.
Steyerberg, E.W et al., “Application of Shrinkage Techniques in Logistic Regression Analysis: A Case Study”, Statistica Neerlandica, 55(1), 2001, 76-88.
Stiller, et al., “Direct Multiplex Sequencing (DMPS)—A Novel Method for Targeted High-thoroughput Sequencing of Ancient and Highly Degraded DNA”, Genome Research, vol. 19, No. 10, Jul. 27, 2009, 1843-1848.
Stolerman, Elliot S. et al., “Haplotype structure of the ENPP1 Gene and Nominal Association of the K121Q missense single nucleotide polymorphism with glycemic traits in the Framingham Heart Study”, Diabetes, vol. 57, Issue 7, Jul. 1, 2008, 1971-1977.
Strom, C. et al., “Three births after preimplantation genetic diagnosis for cystic fibrosis with sequential first and second polar body analysis”, American Journal of Obstetrics and Gynecology, 178 (6), 1998, 1298-1306.
Strom, Charles M. et al., “Neonatal Outcome of Preimplantation Genetic Diagnosis by Polar Body Removal: The First 109 Infants”, Pediatrics, 106(4), 2000, 650-653.
Stroun, Maurice et al., “Prehistory of the Notion of Circulating Nucleic Acids in Plasma/Serum (CNAPS): Birth of a Hypothesis”, Ann. NY. Acad. Sci., 1075, 2006, 10-20.
Su, S.Y. et al., ““Inferring combined CNV/SNP haplotypes from genotype data””, Bioinformatics, vol. 26, No. 11,1, Jun. 1, 2010, 1437-1445.
Su, Z. et al., “A Platform for Rapid Detection of Multiple Oncogenic Mutations With Relevance to Targeted Therapy in Non—Small-Cell Lung Cancer”, The Journal of Molecular Diagnostics,, vol. 13, No. 1, Jan. 2011, 74-84.
Sun, Guihua et al., “SNPs in human miRNA genes affect biogenesis and function”, RNA, 15(9), 2009, 1640-1651.
Swarup, V. et al., “Circulating (cell-free) nucleic acids—A promising, non-invasive tool for early detection of several human diseases”, FEBS Letters, vol. 581, 2007, 795-799.
Sweet-Kind Singer, J. A. et al., “Log-penalized linear regression”, IEEE International Symposium on Information Theory, 2003. Proceedings, 2003, 286.
Swinkels, D. W. et al., “Effects of Blood-Processing Protocols on Cell-free DNA Quantification in Plasma”, Clinical Chemistry, vol. 49, No. 3, 2003, 525-526.
Syvanen, A.C., “Toward genome-wide SNP genotyping”, Nature Genetics Supplement, vol. 37, Jun. 2005, S5-S10.
Taback, B. et al., “Quantification of Circulating DNA in the Plasma and Serum of Cancer Patients”, Ann. N.Y. Acad. Sci, vol. 1022, 2004, 17-24.
Takala, et al., “A High-throughput Method for Quantifying Alleles and Haplotypes of the Malaria Vaccine Candidate Plasmodium Falciparum Merozoite Surface Protein-1 19 kDa”, Malaria Journal, vol. 5:31, Apr. 20, 2006, 1-10.
Takano, T. et al., “Epidermal Growth Factor Receptor Gene Mutations and Increased Copy Numbers Predict Gefitinib Sensitivity in Patients With Recurrent Non-Small-Cell Lung Cancer”, Journal of Clinical Oncology, vol. 23, No. 28, Oct. 1, 2005, 6829-6837.
Takara Biomedicals, “Competitive PCR Guide”, Lit. # L0126, Aug. 1999, 9 pages.
Takashima, Y. et al., “Expansion-contraction of photoresponsive artificial muscle regulated by host-guest interactions”, Nature Communications, vol. 3, No. 1270, Dec. 11, 2012, 8 pages.
Taliun, D. et al., “Efficient haplotype block recognition of very long and dense genetic sequences”, BMC Bioinformatics, vol. 15 (10), 2014, 1-18.
Tamura, et al., “Sibling Incest and formulation of paternity probability: case report”, Legal Medicine, 2000, vol. 2, p. 189-196.
Tang, et al., , Multiplex fluorescent PCR for noninvasive prenatal detection of fetal-derived paternally inherited diseases using circulatory fetal DNA in maternal plasma, Eur J Obstet Gynecol Reprod Biol, 2009, v.144, No. 1, p. 35-39.
Tang, N. et al., “Detection of Fetal-Derived Paternally Inherited X-Chromosome Polymorphisms in Maternal Plasma”, Clinical Chemistry, 45 (11), 1999, 2033-2035.
Tebbutt, S. J. et al., “Microarray genotyping resource to determine population stratification in genetic association studies of complex disease”, BioTechniques, vol. 37, Dec. 2004, 977-985.
Ten Bosch, J., “Keeping Up With the Next Generation Massively Parallel Sequencing in Clinical Diagnostics”, Journal of Molecular Diagnostics, vol. 10, No. 6, 2008, 484-492.
Tewhey, R. et al., “Microdroplet-based PCR enrichment for large-scale targeted sequencing”, Nature Biotechnology, vol. 27, No. 11, Nov. 2009, 1025-1031.
Tewhey, R. et al., “The importance of phase information for human genomics”, Nature Reviews Genetics, vol. 12, No. 3, Mar. 1, 2011, 215-223.
Thavarajah, R. et al., “Chemical and physical basics of routine formaldehyde fixation”, Journal of Oral and Maxillofacial Pathology, vol. 16, No. 3, 2012, 400-405.
The Bump Message Boards, The Bump (Panorama Test, attached), Jul. 1, 2013, 8 pages.
The International Hapmap Consort, “The International HapMap Project”, Nature, vol. 426, Dec. 18, 2003, 789-796.
Thermofisher Scientific, “Ion AmpliSeq Cancer Hotspot Panel v2”, Retrieved from the Internet: https://tools.thermofisher.com/content/sfs/brochures/Ion-AmpliSeq-Cancer-Hotspot-Panel-Flyer.pdf, 2015, 2 pages.
Thomas, M.R et al., “The Time of Appearance and Disappearance of Fetal DNA from the Maternal Circulation”, Prenatal Diagnosis, 15, 1995, 641-646.
Thompson, J. C. et al., “Detection of Therapeutically Targetable Driver and Resistance Mutations in Lung Cancer Patients by Next-Generation Sequencing of Cell-Free Circulating Tumor DNA”, Clin Cancer Res, vol. 22, No. 23, Dec. 1, 2016, 5772-5782.
Thornton, Brenda et al., “Real-time Pcr (qPCR) Primer Design Using Free Online Software”, Biochemistry and Molecular Biology Education, vol. 39, 2011, pp. 145-154.
Tiersch, T. R. et al., “Reference Standards for Flow Cytometry and Application in Comparative Studies of Nuclear DNA Content”, Cytometry, vol. 10, Mar. 21, 1989, 706-710.
Tong, et al., “Diagnostic Developments Involving Cell-free (Circulating) Nucleic Acids”, Clinica Chimica Acta, vol. 363, No. (1-2), Aug. 26, 2005, 187-196.
Tong, Yu et al., “Noninvasive Prenatal Detection of Fetal Trisomy 18 by Epigenetic Allelic Ratio Analysis in Maternal Plasma: Theoretical and Empirical Considerations”, Clinical Chemistry, 52(12), 2006, 2194-2202.
Tong, Yu K. et al., “Noninvasive Prenatal Detection of Trisomy 21 by Epigenetic-Genetic Chromosome-Dosage Approach”, Clinical Chemistry, 56(1), 2010, 90-98.
Toshikazu, et al., “Estimation of Haplotype Frequencies, Linkage-disequilibrium Measures, and Combination of Haplotype Copies in Each Pool by Use of Pooled DNA Data”, American Journal of Human Genetics, vol. 72, Jan. 17, 2003, 384-398.
Tounta, G et al., “Non-invasive prenatal diagnosis using cell-free fetal nucleic acids in maternal plasma: Progress overview beyond predictive and personalized diagnosis”, EPMA Journal, vol. 2, Issue 2, 2011, 163-171.
Tounta, G. et al., “A Multiplex PCR for Non-invasive Fetal RHD Genotyping Using Cell-free Fetal DNA”, in vivo, vol. 25, 2011, 411-418.
Treff, N. R. et al., “Single Cell Whole Genome Amplification Technique Significantly Impacts the Accuracy and Precision of Microarray Based 23 Chromosome Aneuploidy Screening”, Poster Presentations Preimplantation Genetic Diagnosis, vol. 88, Supplement 1, Sep. 1, 2007, S231.
Troeger, C. et al., “Approximately Half of The Erythroblasts in Maternal Blood are of Fetal Origin”, Molecular Human Reproduction, vol. 5, No. 12, Dec. 1, 1999, 1162-1165.
Troutt, et al., “Ligation-anchored PCR: A Simple Amplification Technique with Single-sided Specificity”, Proceedings of the National Academy of Sciences, vol. 89, Oct. 1992, 9823-9825.
Troyanskaya, Olga G. et al., “A Bayesian Framework for Combining Heterogeneous Data Sources for Gene Function Prediction (in Saccharomyces cerevisiae)”, PNAS, 100(14), 2003, 8348-8353.
Tsang, Jason C. et al., “Circulating Nucleic Acids in Plasma/Serum”, Pathology, vol. 39, No. 2, Apr. 1, 2007, 197-207.
Tsangaris, G. T. et al., “Proteomic analysis of amniotic fluid in pregnancies with Down syndrome”, Proteomics, vol. 6, 2006, 4410-4419.
Tseng, Jeng-Sen et al., “Dynamic Plasma EGFR Mutation Status as a Predictor of EGFR-TKI Efficacy in Patients with fGFR-Mutant Lung Adenocarcinoma”, Thorac Oncol., vol. 10, 2015, 603-610.
Tsui, N. B. et al., “Systematic micro-array based identification of placental mRNA in maternal plasma: towards non-invasive prenatal gene expression profiling”, J. Med. Genet, vol. 41, 2004, 461-467.
Tsui, Nancy B.Y et al., “Non-Invasive Prenatal Detection of Fetal Trisomy 18 by RNA-SNP Allelic Ratio Analysis Using Maternal Plasma SERPINB2 mRNA: A Feasibility Study”, Prenatal Diagnosis, 29, 2009, 1031-1037.
Tu, J. et al., “Pair-barcode high-throughput sequencing for large-scale multiplexed sample analysis”, BMC Genomics, vol. 13, No. 43, Jan. 25, 2012, 1-9.
Tufan, N L. et al., “Analysis of Cell-Free Fetal DNA from Maternal Plasma and Serum Using a Conventional Multiplex PCR: Factors Influencing Success”, The Turkish Journal of Medical Sciences, vol. 35, 2005, 85-92.
Turner, E. et al., “Massively Parallel Exon Capture and Library-Free Resequencing Across 16 Genomes”, Nature Methods, 6 (5), 2009, 315-316.
Tuzcu, et al., “Intravascular Ultrasound Evidence of Angiographically Silent Progression in Coronary Atherosclerosis Predicts Long-term Morbidity and Mortality After Cardiac Transplantation”, The American Journal of Cardiology, vol. 45, No. 9, May 3, 2005, 1538-1542.
Tynan, J. A. et al., “Restriction Enzyme-Mediated Enhanced Detection of Circulating Cell-Free Fetal DNA in Maternal Plasma”, The Journal of Molecular Diagnostics, vol. 13, No. 4, Jul. 2011, 382-389.
Tzimagiorgis, G. et al., “Recovering circulating extracellular or cell-free RNA from bodily fluids”, Cancer Epidemiology, vol. 35, 2011, 580-589.
Umetani, N. et al., “Increased Integrity of Free Circulating DNA in Sera of Patients with Colorectal or Periampullary Cancer: Direct Quantitative PCR for ALU Repeats”, Clinical Chemistry, vol. 52, No. 6, 2006, 1062-1069.
Urbaniak, S. J. et al., “RhD haemolytic disease of the fetus and the newborn”, Blood Reviews, vol. 14, 2000, 44-61.
Urbanova, M. et al., “Circulating Nucleic Acids as a New Diagnostic Tool”, Cellular & Molecular Biology Letters, vol. 15, 2010, 242-259.
Vallone, P. M. et al., “A multiplex allele-specific primer extension assay for forensically informative SNPs distributed throughout the mitochondrial genome”, Int J Legal Medicine, vol. 118, Feb. 4, 2004, 147-157.
Vallone, Peter, “AutoDimer: a Screening Tool for Primer-Dimer and Hairpin Structures”, Bio Techniques, 37, 2004, 226-231.
Vallone, Peter M. et al., “Demonstration of Rapid Multiplex PCR Amplification Involving 16 Genetic Loci”, Forensic Science International: Genetics, vol. 3, 2008, pp. 42-45.
Van Den Oever, J. M. et al., “Single Molecule Sequencing of Free DNA from Maternal Plasma for Noninvasive Trisomy 21 Detection”, Clinical Chemistry, vol. 58, No. 4, 2012, 699-706.
Van Uitert, I. et al., “The influence of different membrane components on the electrical stability of bilayer lipid membranes”, Biochimica et Biophysica Acta, vol. 1798, 2010, 21-31.
Vanneste, Marion et al., “Functional Genomic Screening Independently Identifies CUL3 as a Mediator of Vemurafenib Resistance via Src-RAC1 Signaling Axis”, Frontiers in Oncology, vol. 10, 2020, 16 pages.
Varley, Katherine Elena et al., “Nested Patch PCR Enables Highly Multiplexed Mutation Discovery in Candidate Genes”, Genome Res., 18(11), 2008, 1844-1850.
Verlaan, et al., “Allele-specific Chromatin Remodeling in the ZPBP22/GSDMB/ORMDL3 Locus Associated with the Risk of Asthma and Autoimmune Disease”, The American Journal of Human Genetics, vol. 85, No. 3, Sep. 11, 2009, 377-393.
Verlaan, et al., “Targeted Screening of Cis-Regulatory Variation in Human Haplotypes”, Genome Research, vol. 19, No. 1, Jan. 1, 2009, 118-127.
Verlinsky, Y. et al., “Over a Decade of Experience with Preimplantation Genetic Diagnosis”, Fertility and Sterility, 82 (2), 2004, 302-303.
Vlaminck, I. D. et al., “Circulating Cell-Free DNA Enables Noninvasive Diagnosis of Heart Transplant Rejection”, Sci Transl Med., vol. 6, No. 241, Jun. 18, 2018, 26 pages.
Voelkerding, et al., “Next-generation Sequencing: From Basic Research to Diagnostics”, Clinical Chemistry, vol. 55, No. 4, Apr. 1, 2009, 641-658.
Vogelstein, B. et al., “Digital PCR”, Proc. Natl. Acad. Sci. USA, vol. 96, Aug. 1999, 9236-9241.
Von Ahsen, Nicolas et al., “Oligonucleotide Melting Temperatures under PCR Conditions: Nearest-Neighbor Corrections for Mg2+, Deoxynucleotide Triphosphate, and Dimethyl Sulfoxide Concentrations with Comparison to Alternative Empirical Formulas”, Clinical Chemistry, vol. 47, 2001, pp. 1956-1961.
Von Eggeling, F. et al., “Applications of Random PCR”, Cellular and Molecular Biology, vol. 41, No. 5, 1995, 653-670.
Wagner, Jasenka et al., “Non-Invasive Prenatal Paternity Testing from Maternal Blood”, Int. J. Legal Med., 123, 2009, 75-79.
Wang, D. G. et al., “Large-Scale Identification, Mapping, and Genotyping of Single-Nucleotide Polymorphisms in the Human Genome”, Science, vol. 280, May 15, 1998, 1077-1082.
Wang, Eric et al., “Gestational Age and Maternal Weight Effects on Fetal Cell-Free DNA in Maternal Plasma”, Prenatal Diagnosis, 33, 2013, 662-666.
Wang, Hui-Yun et al., “A genotyping system capable of simultaneously analyzing >1000 single nucleotide polymorphisms in a haploid genome”, Genome Res., 15, 2005, 276-283.
Wang, J. et al., “Genome-wide Single-Cell Analysis of Recombination Activity and De Novo Mutation Rates in Human Sperm”, Cell, vol. 150, Jul. 20, 2012, 402-412.
Wang, S. et al., “Potential Clinical Significance of a Plasma-Based KRAS Mutation Analysis in Patients with Advanced Non-Small Cell Lung Cancer”, Clin Cancer Res, vol. 16, No. 4, Feb. 15, 2010, 1324-1330.
Wang, T.L. et al., “Digital karyotyping”, PNAS, vol. 99, No. 25, Dec. 10, 2002, 16156-16161.
Wang, W.-P. et al., “Multiplex single nucleotide polymorphism genotyping by adapter ligation-mediated allele-specific amplification”, Analytical Biochemistry, vol. 355, May 5, 2006, 240-248.
Wang, Yuker et al., “Allele quantification using molecular inversion probes (MIP)”, Nucleic Acids Research, vol. 33, No. 21, Nov. 28, 2005, 14 pgs.
Wapner, R. et al., “Chromosomal Microarray Versus Karyotyping for Prenatal Diagnosis”, The New England Journal of Medicine, 367 (23), 2012, 2175-2184.
Wapner, R. et al., “First-Trimester Screening for Trisomies 21 and 18”, The New England Journal of Medicine, vol. 349, No. 15, Oct. 9, 2003, 1405-1413.
Wapner, R. J. et al., “Expanding the scope of noninvasive prenatal testing: detection of fetal microdeletion syndromes”, American Journal of Obstetrics & Gynecology, vol. 212, Dec. 17, 2014, 1.e1-1.e9.
Wartell, Roger M. et al., “Thermal Denaturation of DNA Molecules: A Comparison of Theory with Experiment”, Physics Reports, vol. 126, 1985, pp. 67-107.
Wasson, Jon et al., “Assessing Allele Frequencies of Single Nucleotide Polymorphisms in DNA Pools by Pyrosequencing Technology”, BioTechniques, vol. 32, No. 5, May 1, 2002, 1144-1152.
Watkins, N. et al., “Thermodynamic contributions of single internal rA ⋅dA, rC ⋅ dC, rG ⋅ dG and rU ⋅ dT mismatches in RNA/DNA duplexes”, Nucleic Acids Research, 9 (5), 2010, 1894-1902.
Watt, Heather L., “Sex Diagnosis of Preimplantation Porcine Embryos through PCR Amplification of the Sry Gene”, Sex Diagnosis of Preimplantation Porcine Embryos Through PCR Amplification of the SRY Gene (1998) (“Watt (1998)”), 1998, 151 pages.
Wei, C. et al., “Detection and Quantification by Homogeneous PCR of Cell-free Fetal DNA in Maternal Plasma”, Clinical Chemistry, vol. 47, No. 2, 2001, 336-338.
Wei, Ting et al., “Novel Approaches to Mitigate Primer Interaction and Eliminate Inhibitors in Multiplex PCR, Demonstrated Using an Assay for Detection of three Strawberry Viruses”, Journal of Virological Methods, vol. 151, 2008, pp. 132-139.
Weiss, C. A., “Chapter 8: Confidence Intervals for One Population Mean”, Introductory Statistics, Sixth Edition, 2002, 340-381.
Wellnhofer, et al., “Angiographic Assessment of Cardiac Allograft Vasculopathy: Results of a Consensus Conference of the Task Force for Thoracic Organ Transplantation of the German Cardiac Society”, Transplant International, vol. 23, No. 11, Aug. 19, 2010, 1094-1104.
Wells, D, “Microarray for Analysis and Diagnosis of Human Embryos”, 12th International Congress on Prenatal Diagnosis and Therapy, Budapest, Hungary, 2004, 9-17.
Wells, Dagan, “Advances in Preimplantation Genetic Diagnosis”, European Journal of Obstetrics and Gynecology and Reproductive Biology, 115S, 2004, S97-S101.
Wells, Dagan, “Detailed Chromosomal and Molecular Genetic Analysis of Single Cells by Whole Genome Amplification and Comparative Genomic Hybridisation”, Nucleic Acids Research, 27, 4, 1999, 1214-1218.
Wen, Daxing et al., “Universal Multiples PCR: A Novel Method of Simultaneous Amplification of Multiple DNA Fragments”, Plant Methods, 8(32), Null, 2012, 1-9.
What to Expect Message Boards, , What to Expect (Weird Harmony results), May 1, 2015, 7 pages.
Widlak, P. et al., “Cleavage Preferences of the Apoptotic Endonuclease DFF 40 (Caspase˜activated DNase or Nuclease) on Naked DNA and Chromatin Substrates”, The Journal of Biological Chemistry, vol. 275, No. 11, Mar. 17, 2000, 8228-8232.
Wiedmann, Ralph T. et al., “SNP Discovery in Swine by Reduced Representation and High Throughput Pyrosequencing”, BMC Genetics, vol. 9, Article No. 81, Dec. 4, 2008, 1-7.
Wikipedia, “Buffy coat”, Retrieved from “https://en.wikipedia.orgJw/index.php?title=Buffy_coat&oldid=900992886 ”, Jun. 9, 2019, 2 pgs.
Wikipedia, “Maximum a posteriori estimation”, https://en.wikipedia.org/w/index.php?title=Maximum_a_posteriori_estimation&oldid=26878808, [retrieved on Aug. 1, 2017], Oct. 30, 2005, 2 pages.
Wikipedia, “Stimulant”, 2016, 17 pages.
Wilkening, Stefan et al., “Determination of Allele Frequency in Pooled DNA: Comparison of Three PCR-based Methods”, Bio Techniques, vol. 39, No. 6, May 30, 2005, 853-857.
Wilkinson, Sarah T. et al., “Decreased MHC Class II Expression in Diffuse Large B-Cell Lymphoma does not Correlate with CPG Methylation of Ciita Promoters III and IV”, Leuk Lymphoma, vol. 50, 2009, pp. 1875-1878.
Wilton, et al., “Birth of a Healthy Infant After Preimplantation Confirmation of Euploidy by Comparative Genomic Hybridization”, N. Engl. J. Med., 345(21), 2001, 1537-1541.
Wilton, L., “Preimplantation Genetic Diagnosis and Chromosome Analysis of Blastomeres Using Comparative Genomic Hybridization”, Human Reproduction Update, 11 (1), 2005, 33-41.
Winsor, E. J. et al., “Maternal Cell Contamination in Uncultured Amniotic Fluid”, Prenatal Diagnosis, vol. 16, 1996, 49-54.
Witherspoon, David J. et al., “Mobile Element Scanning (Me-scan) by Targeted High-throughput Sequencing”, BMC Genomics, vol. 410, 2010, 15 pages.
Wittwer, C. T. et al., “Real-Time Multiplex PCR Assays”, Methods, vol. 25, 2001, 430-448.
Wong, K. H. et al., “Multiplex Illumina Sequencing Using DNA Barcoding”, Current Protocols in Molecular Biology, vol. 101, Jan. 2013, 7.11.1-7.11.11.
Wong, K. K. et al., “Allelic imbalance analysis by high-density single nucleotide polymorphic allele (SNP) array with whole genome amplified DNA”, Nucleic Acids Research, vol. 32, No. 9, May 17, 2004, 8 pages.
Wright, C. et al., “The use of cell-free fetal nucleic acids in maternal blood for non-invasive prenatal diagnosis”, Human Reproduction Update, vol. 15, No. 1, 2009, 139-151.
Wright, C. F. et al., “Cell-free fetal DNA and RNA in maternal blood: implications for safer antenatal testing”, BMJ, vol. 39, Jul. 18, 2009, 161-165.
Wright, Caroline et al., “Cell-free Fetal Nucleic Acids for Noninvasive Prenatal Diagnosis”, PHG Foundation, Jan. 1, 2009, 1-64.
Wu, T.L. et al., “Cell-free DNA: measurement in various carcinomas and establishment of normal reference range”, Clinica Chimica Acta, vol. 321, 2002, 77-87.
Wu, Y. Y. et al., “Rapid and/or high-throughput genotyping for human red blood cell, platelet and leukocyte antigens, and forensic applications”, Clinica Chimica Acta, vol. 363, 2006, 165-176.
Xia, et al., “Simultaneous Quantitative Assessment of Circulating Cell-free Mitochondrial and Nuclear DNA by Multiplex Real-time PCR”, Genetics and Molecular Biology, vol. 32, No. 1, Mar. 1, 2009, 20-24.
Xia, Tianbing et al., “Thermodynamic Parameters for an Expanded Nearest-Neighbor Model for Formation of RNA Duplexes with Watson-Crick Base Pairs”, Biochemistry, 37, 1998, 14719-14735.
Xian, et al., “Advances on Circulating Fetal DNA in Maternal Plasma”, Chinese Medical Journal, vol. 120, No. 14, Jul. 2, 2007, 1256-1259.
Xie, et al., “CNV-SEQ, A New Method to Detect Copy Number Variation Using Highthroughput Sequencing”, BMC Bioinformatics, vol. 10:80, Mar. 6, 2009, 1-9.
Xu, N. et al., “A Mutation in the Fibroblast Growth Factor Receptor 1 Gene Causes Fully Penetrant Normosmic Isolated Hypogonadotropic Hypogonadism”, The Journal of Clinical Endocrinology & Metabolism, vol. 92, No. 3, 2007, 1155-1158.
Xu, S. et al., “Circulating tumor DNA identified by targeted sequencing in advanced-stage non-small cell lung cancer patients”, Cancer Letters, vol. 370, 2016, 324-331.
Xu, W. et al., “A Novel Universal Primer-Multiplex-PCR Method with Sequencing Gel Electrophoresis Analysis”, PLOS One, vol. 7, No. 1, Jan. 17, 2012, 10 pgs.
Xue, et al., “Optimizing the Yield and Utility of Circulating Cell-free DNA From Plasma and Serum”, Clinica Chimica Acta, vol. 404, No. 2, Jun. 27, 2009, 100-104.
Yamada, T. et al., “Detection of K-ras Gene Mutations in Plasma DNA of Patients with Pancreatic Adenocarcinoma: Correlation with Clinicopathological Features”, Clinical Cancer Research, vol. 4, Jun. 1998, 1527-1532.
Yamada, T. et al., “PrimerStation: a highly specific multiplex genomic PCR primer design server for the human genome”, Nucleic Acids Research, vol. 34, 2006, W665-W669.
Yang, Lin et al., “64-MDCT Coronary Angiography of Patients With Atrial Fibrillation: Influence of Heart Rate on Image Quality and Efficacy in Evalution of Coronary Artery Disease”, AJR, vol. 193, No. 3, Sep. 1, 2009, 795-801.
Yaron, Y., “The implications of non-invasive prenatal testing failures: a review of an under-discussed phenomenon”, Prenatal Diagnosis, vol. 36, 2016, 391-396.
Yeh, Iwei et al., “Knowledge Acquisition, Consistency Checking and Concurrency Control for Gene Ontology (GO)”, Bioinformatics, 19, 2, 2003, 241-248.
Yijen, et al., “Noninvasive Evaluation of Cardiac Allograft Rejection by Cellular and Functional Cardiac Magnetic Resonance”, JACC: Cardiovacular Imaging, vol. 2, No. 6, Jun. 1, 2009, 731-741.
Yilmaz, A. et al., “Comparative Evaluation of Left and Right Ventricular Endomyocardial Biopsy”, Circulation, vol. 122, No. 9, Aug. 31, 2010, 900-909.
You, Frank M. et al., “BatchPrimer3: A high throughput web application for PCR and sequencing primer design”, BMC Bioinformatics, Biomed Central, London, GB, vol. 9, No. 1, May 29, 2008 (May 29, 2008), p. 253.
Yuan, X. et al., “Probability Theory-based SNP Association Study Method for Identifying Susceptibility Loci and Genetic Disease Models in Human Case-Control Data”, IEEE Trans Nanobioscience, vol. 9, No. 4, Dec. 2010, 232-241.
Yuanxin, Yan et al., “T-linker-specific Ligation PCR (T-linker Pcr): An Advanced PCR Technique for Chromosome Walking or for Isolation of Tagged DNA Ends”, Nucleic Acids Research, vol. 31, No. 12, e68, 2003, 7 pages.
Yung, T. K. et al., “Single-Molecule Detection of Epidermal Growth Factor Receptor Mutations in Plasma by Microfluidics Digital PCR in Non-Small Cell Lung Cancer Patients”, Clinical Cancer Research, vol. 15, Mar. 10, 2009, 2076-2084.
Zachariah, R. et al., “Circulating cell-free DNA as a potential biomarker for minimal and mild endometriosis”, Reproductive BioMedicine Online, vol. 18, No. 3, Jan. 27, 2009, 4007-411.
Zhang, et al., “Diagnosis of Acute Rejection by Analysis of Urinary DNA of Donor Origin in Renal Transplant Recipients”, Transplantation Proceedings, vol. 33, No. 1-2, Feb. 2001, 380-381.
Zhang, et al., “Use of PCR And PCR-SSP for Detection of Urinary Donor-Origin Dna in Renal Transplant Recipients With Acute Rejection”, Chinese Medical Journal, vol. 116, No. 2, Feb. 2003, 191-194.
Zhang, J. et al., “Presence of Donor-and Recipient-derived DNA in Cell-free Urine Samples of Renal Transplantation Recipients: Urinary DNA Chimerism”, Clinical Chemistry, vol. 45, No. 10, 1999, 1741-1746.
Zhang, Kun et al., “Digital RNA Alleotyping Reveals Tissue-specific and Allele-specific Gene Expression in Human”, Nature Methods, vol. 6, No. 8, Jul. 20, 2009, 613-618.
Zhang, L. et al., “Whole genome amplification from a single cell: Implications for genetic analysis”, Proc. Nat'l. Acad. Sci. USA, vol. 89, Jul. 1992, 5847-5851.
Zhang, Rui et al., “Quantifying RNA allelic ratios by microfluidic multiplex PCR and sequencing”, Nature Methods, 11(1), 2014, 51-56.
Zhao, et al., “Urinary Thromboxane B2 in Cardiac Transplant Patients as a Screening Method of Rejection”, Prostaglandins, vol. 54, No. 6, Dec. 1, 1997, 881-889.
Zhao, Xiaojun et al., “An Integrated View of Copy Number and Allelic Alterations in the Cancer Genome Using Single Nucleotide Polymorphism Arrays”, Cancer Research, 64, 2004, 3060-3071.
Zheng, S. et al., “Whole Genome Amplification Increases the Efficiency and Validity of Buccal Cell Genotyping in Pediatric Populations1”, Cancer Epidemiology, Biomarkers & Prevention, vol. 10, Jun. 2001, 697-700.
Zheng, Z et al., “Anchored Multiplex PCR for Targeted Next-generation Sequencing”, Nature Medicine, vol. 20, No. 12, Dec. 2014, 1479-1486.
Zhong, X Y. et al., “Detection of Fetal Rhesus D and Sex Using Fetal DNA from Maternal Plasma by Multiplex Polymerase Chain Reaction”, British Journal of Obstetrics and Gynaecology, vol. 107, Jun. 2000, 766-769.
Zhong, X. et al., “Risk free simultaneous prenatal identification of fetal Rhesus D status and sex by multiplex real-time PCR using cell free fetal DNA in maternal plasma”, Swiss Medical Weekly, vol. 131, Mar. 2001, 70-74.
Zhong, Xiao Y. et al., “Cell-free DNA in Urine: A Marker for Kidney Graft Rejection, but Not for Prenatal Diagnosis ?”, Annals of the New York Academy of Sciences, vol. 945, Sep. 1, 2001, 250-257.
Zhou, et al., “Pyrosequencing, A High-throughput Method for Detecting Single Nucleotide Polymorphisms in the Dihydrofolate Reductase and Dihydropteroate Synthetase Genes of Plasmodiym Falciparum”, Journal of Clinical Microbiology, vol. 44, No. 11, Nov. 1, 2006, 3900-3910.
Zhou, W. et al., “Counting Alleles Reveals a Connection Between Chromosome 18q Loss and Vascular Invasion”, Nature Biotechnology, 19, 2001, 78-81.
Zhou, W. et al., “Counting alleles to predict recurrence of early-stage colorectal cancers”, The Lancet, vol. 359, Jan. 19, 2002, 219-225.
Zimmer, et al., “Transplant Coronary Artery Disease”, JACC: Cardiovascular Interventions, vol. 3, No. 4, Apr. 1, 2010, 367-377.
Zimmermann, et al., “Noninvasive Prenatal Aneuploidy Testing of Chromosomes 13, 18, 21 X, and Y, Using targeted Sequencing of Polymorphic Loci”, Prenatal Diagnosis, 32, 2012, 1-9.
Zimmermann, B., “Declaration Under 37 CFR 1.32”, filed in U.S. Appl. No. 14/171,587, filed Feb. 3, 2014, 4 pgs.
Zimmermann, B. et al., “Digital PCR: a powerful new tool for noninvasive prenatal diagnosis?”, Prenatal Diagnosis, vol. 28, Nov. 10, 2008, 1087-1093.
Zimmermann, B., “Noninvasive prenatal aneuploidy testing of chromosomes 13, 18, 21, X, and Y, using targeted sequencing of polymorphic loci, Supplemental Information”, Prenatal Diagnosis, vol. 32, 2012, 7 pages.
Zimmermann, B. et al., “Novel Real-Time Quantitative PCR Test for Trisomy 21”, Clinical Chemistry, vol. 48, No. 2, 2002, 362-363.
Zimmermann, B. et al., “Optimized Real-Time Quantitative PCR Measurement of Male Fetal DNA in Maternal Plasma”, Clinical Chemistry, vol. 51, No. 9, 2005, 1598-1604.
Zimmermann, B. et al., “Real-Time Quantitative Polymerase Chain Reaction Measurement of Male Fetal DNA in Maternal Plasma”, Methods in Molecular Medicine, vol. 132, 2007, 43-49.
Zimmermann, B. et al., “Use of Real-Time Polymerase Chain Reaction for the Detection of Fetal Aneuploidies”, Methods in Molecular Biology, vol. 336, Feb. 2006, 83-100.
Zlotogora, J. , “Penetrance and expressivity in the molecular age”, Genetics in Medicine, vol. 5, No. 5, 2003, 347-352.
Related Publications (1)
Number Date Country
20210269879 A1 Sep 2021 US
Provisional Applications (1)
Number Date Country
62599011 Dec 2017 US