WO2015069933A1 - Adn acellulaire circulant pour le diagnostic du rejet de greffe - Google Patents

Adn acellulaire circulant pour le diagnostic du rejet de greffe Download PDF

Info

Publication number
WO2015069933A1
WO2015069933A1 PCT/US2014/064402 US2014064402W WO2015069933A1 WO 2015069933 A1 WO2015069933 A1 WO 2015069933A1 US 2014064402 W US2014064402 W US 2014064402W WO 2015069933 A1 WO2015069933 A1 WO 2015069933A1
Authority
WO
WIPO (PCT)
Prior art keywords
transplant
donor
nucleic acids
rejection
dna
Prior art date
Application number
PCT/US2014/064402
Other languages
English (en)
Inventor
Stephen R. Quake
Original Assignee
The Board Of Trustees Of The Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The Board Of Trustees Of The Leland Stanford Junior University filed Critical The Board Of Trustees Of The Leland Stanford Junior University
Publication of WO2015069933A1 publication Critical patent/WO2015069933A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • the present invention generally relates to utilizing circulating cell-free nucleic acid in order to diagnose the rejection or acceptance of a transplant.
  • endomyocardial biopsy is the gold standard for cardiac allograft monitoring, but requires an expensive and invasive procedure.
  • the endomyocardial biopsy is an expensive and invasive procedure that is limited by sampling error and interobserver variability in grading.
  • cardiac biopsies may cause patient discomfort and rare but serious complications, including arterial puncture, arrhythmias, conduction abnormalities, biopsy induced tricuspid regurgitation, and even cardiac perforation.
  • Methods of the invention provide a non-invasive diagnostic method for monitoring the success of an implant post-implantation with comparable predictive values of biopsies.
  • Methods of the invention involve quantification of donor-derived circulating cell-free nucleic acid, such as DNA (cfdDNA), in order to determine the success of an implant. Quantification can be achieved through high throughput sequencing of the cfdDNA.
  • the invention provides methods of diagnosing or predicting transplant status or outcome comprising the steps of: (i) providing a sample from a subject who has received a transplant from a donor; (ii) determining the presence or absence of one or more nucleic acids from the donor transplant, where the one or more nucleic acids from the donor are identified based on a predetermined marker profile; (iii) performing bioinformatics analysis to improve analysis quality and (iv) diagnosing or predicting transplant status or outcome based on the presence or absence of the one or more nucleic acids.
  • the sample for analysis is taken at least two weeks following transplantation. In some embodiments the sample for analysis is take at least 4 months following transplantation, e.g. about 4, about 5, about 6, about 7, about 8, about 9, about 10, about 1 1 , about 12, about 13, about 14, about 15, about 20, about 24 or more months following transplantation.
  • methods of the invention involve detecting a sample level of circulating nucleic acid in a biological sample from a transplant recipient, determining a donor level of circulating nucleic acid in the sample corresponding to the donor of the transplant; and comparing the donor level of circulating nucleic acid to a reference level of nucleic acid.
  • the method further provides for determining whether a difference exists between the donor level and the reference level, and characterizing the transplant as rejected if a difference is detected.
  • the donor transplant is a lung transplant or a heart transplant.
  • the transplant status or outcome comprises rejection, tolerance, non-rejection based allograft injury, transplant function, transplant survival, chronic transplant is injury, or titer pharmacological immunosuppression.
  • the non-rejection based allograft injury is selected from the group of ischemic injury, virus infection, peri-operative ischemia, reperfusion injury, hypertension, physiological stress, injuries due to reactive oxygen species and injuries caused by pharmaceutical agents.
  • the sample is selected from the group consisting of blood, serum, urine, and stool.
  • the marker profile is a polymorphic marker profile.
  • the polymorphic marker profile comprises one or more single nucleotide polymorphisms (SNP's), one or more restriction fragment length polymorphisms (RFLP's), one or more short tandem repeats (STRs), one or more variable number of tandem repeats (VNTR's), one or more hypervariable regions, one or more minisatellites, one or more dinucleotide repeats, one or more trinucleotide repeats, one or more tetranucleotide repeats, one or more simple sequence repeats, or one or more insertion elements.
  • the polymorphic marker profile comprises one or more SNPs
  • the marker profile is determined by genotyping the transplant donor.
  • the methods further comprise genotyping the subject receiving the transplant.
  • the methods further comprise establishing a profile of markers, where the markers are distinguishable between the transplant donor and the subject receiving the transplant.
  • the genotyping is performed by a method selected from the group consisting of sequencing, nucleic acid array and PCR.
  • the transplant graft maybe any solid organ and skin transplant.
  • the transplant is selected from the group consisting of kidney transplant, heart transplant, liver transplant, pancreas transplant, lung transplant, intestine transplant and skin transplant.
  • the nucleic acid is selected from the group consisting of double-stranded DNA, single-stranded DNA, single-stranded DNA hairpins, DNA RNA hybrids, RNA and RNA hairpins. In some embodiments, the nucleic acid is selected from the group consisting of double-stranded DNA, single-stranded DNA and cDNA. In some embodiments, the nucleic acid is mRNA. In some embodiments, the nucleic acid is obtained from circulating donor cells. In some embodiments, the nucleic acid is circulating cell-free DNA.
  • the presence or absence of the one or more nucleic acids is determined by a method selected from the group consisting of sequencing, nucleic acid array and PCR.
  • the sequencing is shotgun sequencing.
  • the array is a DNA array.
  • the DNA array is a polymorphism array.
  • the polymorphism array is a SNP array.
  • the methods further comprise quantitating the one or more nucleic acids.
  • the amount of the one or more nucleic acids is indicative of transplant status or outcome.
  • the amount of the one or more nucleic acids above a predetermined threshold value is indicative of a transplant status or outcome.
  • the threshold is a normative value for clinically stable post-transplantation patients with no evidence of transplant rejection or other pathologies.
  • temporal differences in the amount of the one or more nucleic acids are indicative of a transplant status or outcome.
  • the methods described herein have at least 56 % sensitivity.
  • the methods described herein have at least 78 % sensitivity. In some embodiments, the methods described herein have a specificity of about 70% to about 100%. In some embodiments, the methods described herein have a specificity of about 80% to about 100%. In some embodiments, the methods described herein a specificity of about 90% to about 100%. In some embodiments, the methods described herein have a specificity of about 100%.
  • the invention provides computer readable mediums comprising: a set of instructions recorded thereon to cause a computer to perform the steps of: (i) receiving data from one or more nucleic acids detected in a sample from a subject who has received transplant from a donor, where the one or more nucleic acids are nucleic acids from the donor transplant, and where the one or more nucleic acids from the donor are identified based on a predetermined marker profile; and (ii) diagnosing or predicting transplant status or outcome based on the presence or absence of the one or more nucleic acids.
  • the invention provides reagents and kits thereof for practicing one or more of the methods described herein.
  • the concordance of the cfdDNA levels and biopsy data depends on patient age and time from the transplant; it is highest for patients younger than 60 years old and for samples collected greater than about 4 months post-transplant.
  • the non-invasive genome transplant dynamics approach of the invention is a powerful and informative method for routine monitoring of allograft health without incurring the risk, discomfort and expense of an invasive biopsy.
  • Figure 1 Enrollment of patients, collection of clinical samples and analysis workflow.
  • 63 heart transplant recipients were enrolled in the study. Donor and recipient pretransplant whole-blood samples were collected and processed for genotyping. Plasma samples were collected longitudinally post-transplant and circulating cell-free DNA was purified and sequenced. The fraction of donor-derived cell-free DNA was estimated and compared against biopsy scores, when available.
  • FIG. 1 Panels A-D. Principle of the assay and assignment and read statistics. A.
  • the donor and recipient are SNP-genotyped prior to the transplant procedure. Shotgun sequencing of circulating cell-free DNA is performed to calculate the number of donor-derived and recipient-derived DNA molecules. SNP positions for which the donor and recipient are homozygous with a single base present in both alleles allow discrimination of donor and recipient derived sequences (marker n in the cartoon).
  • D Histogram of the number of donor sequence assignments. [0022]
  • Figure 3 Panels A, B. Rate of incorrect donor or recipient sequence assignments.
  • A Histogram of the measured per-sample error rate. The median error rate is 0.04% and 94% of samples had a measured error rate ⁇ 0.15%.
  • B. Linear correlation between the measured donor-DNA fraction and measured error rate (spearman correlation coefficient 0.62). The red line is a linear fit, slope 4.
  • FIG 4 panels A-D. Time dependence of cfdDNA fraction in the absence of rejection, and three examples of acute rejection.
  • B Time course for an adult transplant recipient who suffered from an acute cellular rejection (ACR) episode at month 15 (solid line is fit from panel A).
  • ACR acute cellular rejection
  • C Time course for an adult heart transplant recipient who suffered from an ACR episode (month 9) and subsequently required a new heart transplant (month 10).
  • D Time course for a pediatric heart transplant recipient who suffered from consecutive ACR (months 4 and 12) and antibody-mediated rejection (AMR month 5) episodes.
  • FIG. 5 Panels A-E. Analysis of the performance of cfdDNA as a marker for heart transplant rejection.
  • A. Fraction of cfdDNA for stable heart transplant recipients (biopsy grade 0), recipients diagnosed with mild rejection (1 R/1A ⁇ grade ⁇ 2R/3A) and recipients diagnosed with moderate or severe rejection (grade > 2R/3A), p-values based on Mann- Whitney U test, n denotes the number of samples available for each group.
  • B A receiver- operating characteristic curve tests the performance of donor-derived cfdDNA in classifying rejecting (grade > 2R/3A) and non-rejecting recipients. The area under the curve (AUC) is 0.825.
  • Test performance as a function of the time post-transplant after which samples are taken into account.
  • D Test performance (AUC) as a function of the age of the recipient at the time of transplant (n indicates the number of moderate or severe rejection events recorded for each age group).
  • E Potential for early diagnosis. In many cases the donor DNA level is elevated several months prior to the diagnosis of a moderate-to-severe rejection episode (endomyocardial biopsy grade >2R/3A, red data). Black line, single exponent fit.
  • FIG. 6 depicts the fraction of donor-derived DNA in a biological sample obtained from a lung transplant recipient in the absence of rejection over a course of time.
  • FIG. 7 compares the fraction of donor-derived DNA in a biological sample obtained from a single lung transplant recipient in the absence of rejection over a course of time and a double lung transplant recipient in the absence of rejection over a course of time.
  • FIG. 8 illustrates a change in donor-derived DNA in a biological sample of a lung transplant recipient that is indicative of a rejection.
  • FIG. 9 compares the fraction of donor-derived DNA in a biological sample of lung transplant recipients treated for rejection and patients not treated for rejection.
  • methods of the invention involve detecting a sample level of circulating nucleic acid in a biological sample from a transplant recipient, determining a donor level of circulating nucleic acid in the sample corresponding to the donor of the transplant; and comparing the donor level of circulating nucleic acid to a reference level of nucleic acid.
  • the method further provides for determining whether a difference exists between the donor level and the reference level, and characterizing the transplant as rejected if a difference is detected.
  • transplant status or outcome may comprise rejection, tolerance, non-rejection based transplant injury, transplant function, transplant survival, chronic transplant injury, or titer pharmacological immunosuppression.
  • This invention describes sensitive and non-invasive methods, devices, compositions and kits for monitoring organ transplant patients, and/or for diagnosing or predicting transplant status or outcome (e.g. transplant rejection).
  • the methods, devices, compositions and kits are used to establish a genotype for both the donor and the recipient before transplantation to enable the detection of donor-specific nucleic acids such as DNA or RNA in bodily fluids such as blood or urine from the organ recipient after transplantation.
  • the invention provides methods of determining whether a patient or subject is displaying transplant tolerance.
  • transplant tolerance includes when the subject does not reject a graft organ, tissue or cell(s) that has been introduced into/onto the subject. In other words, the subject tolerates or maintains the organ, tissue or cell(s) that has been transplanted to it.
  • patient or subject as used herein includes humans as well as other mammals.
  • transplant rejection encompasses both acute and chronic transplant rejection.
  • Acute rejection or AR is the rejection by the immune system of a tissue transplant recipient when the transplanted tissue is immunologically foreign. Acute rejection is characterized by infiltration of the transplanted tissue by immune cells of the recipient, which carry out their effector function and destroy the transplanted tissue. The onset of acute rejection is rapid and generally occurs in humans within a few weeks after transplant surgery. Generally, acute rejection can be inhibited or suppressed with immunosuppressive drugs such as rapamycin, cyclosporin A, anti-CD40L monoclonal antibody and the like.
  • Chronic transplant rejection or CR generally occurs in humans within several months to years after engraftment, even in the presence of successful immunosuppression of acute rejection. Fibrosis is a common factor in chronic rejection of all types of organ transplants. Chronic rejection can typically be described by a range of specific disorders that are characteristic of the particular organ.
  • disorders include fibroproliferative destruction of the airway (bronchiolitis obliterans); in heart transplants or transplants of cardiac tissue, such as valve replacements, such disorders include fibrotic atherosclerosis; in kidney transplants, such disorders include, obstructive nephropathy, nephrosclerorsis, tubulointerstitial nephropathy; and in liver transplants, such disorders include disappearing bile duct syndrome.
  • Chronic rejection can also be characterized by ischemic insult, denervation of the transplanted tissue, hyperlipidemia and hypertension associated with immunosuppressive drugs.
  • the invention further includes methods for determining an immunosuppressive regimen for a subject who has received a transplant, e.g., an allograft.
  • Certain embodiments of the invention provide methods of predicting transplant survival in a subject that has received a transplant.
  • the invention provides methods of diagnosing or predicting whether a transplant in a transplant patient or subject will survive or be lost.
  • the invention provides methods of diagnosing or predicting the presence of long-term graft survival.
  • long-term graft survival is meant graft survival for at least about 5 years beyond current sampling, despite the occurrence of one or more prior episodes of acute rejection.
  • transplant survival is determined for patients in which at least one episode of acute rejection has occurred. As such, these embodiments provide methods of determining or predicting transplant survival following acute rejection.
  • Transplant survival is determined or predicted in certain embodiments in the context of transplant therapy, e.g., immunosuppressive therapy, where immunosuppressive therapies are known in the art.
  • methods of determining the class and/or severity of acute rejection are provided.
  • the invention provides methods for diagnosis or prediction of non-rejection based transplant injury.
  • non-rejection based transplant injury include, but are not limited to, ischemic injury, virus infection, peri-operative ischemia, reperfusion injury, hypertension, physiological stress, injuries due to reactive oxygen species and injuries caused by pharmaceutical agents.
  • the transplant organ, tissue or cell(s) may be allogeneic or xenogeneic, such that the grafts may be allografts or xenografts.
  • a feature of the graft tolerant phenotype detected or identified by the subject methods is that it is a phenotype which occurs without immunosuppressive therapy, i.e., it is present in a host that is not undergoing immunosuppressive therapy such that immunosuppressive agents are not being administered to the host.
  • the transplant graft maybe any solid organ and skin transplant.
  • organ transplants that can be analyzed by the methods described herein include but are not limited to kidney transplant, pancreas transplant, liver transplant, heart transplant, lung transplant, intestine transplant, pancreas after kidney transplant, and simultaneous pancreas-kidney transplant.
  • Methods, devices, compositions and kits are provided for diagnosing or predicting transplant status or outcome in a subject who has received a transplant.
  • Circulating, or cell-free, DNA was first detected in human blood plasma in 1948.
  • Circulating DNA has also been useful in healthy patients for fetal diagnostics, with fetal DNA circulating in maternal blood serving as a marker for gender, rhesus D status, fetal aneuploidy, and sex- linked disorders.
  • Fan et al recently demonstrated a strategy for detecting fetal aneuploidy by shotgun sequencing of cell-free DNA taken from a maternal blood sample, a methodology that can replace more invasive and risky techniques such as amniocentesis or chorionic villus sampling.
  • the invention provides non-invasive diagnostics exists for organ transplant patients where sequences from the organ donor, otherwise "foreign" to the patient, can be quantitated specifically. Without intending to be limited to any theory, as cell-free DNA or RNA often arises from apoptotic cells, the relative amount of donor-specific sequences in circulating nucleic acids should provide a predictive measure of on-coming organ failure in transplant patients for many types of solid organ transplantation including, but not limited to, heart, lung, liver, and kidney.
  • the invention provides methods, devices, compositions and kits for detection and/or quantitating circulating nucleic acids, either free in plasma or from circulating cells, for the diagnosis, prognosis, detection and/or treatment of a transplant status or outcome.
  • Example 1 examined gender-mismatched heart transplant recipients and applied digital PCR (Warren, L, Bryder, D., Weissman, I.L., Quake, S.R., Proc Natl Acad Sci, 103, 17807-17812 (2006); Fan, H.C. Quake, S.R., Anal Chem, 79, 7576-7579 (2007)) to detect the level of donor-derived chromosome Y signal in plasma samples taken at the same time that an endomyocardial biopsy determined a grade 3A or 3B rejection episode.
  • digital PCR Warren, L, Bryder, D., Weissman, I.L., Quake, S.R., Proc Natl Acad Sci, 103, 17807-17812 (2006); Fan, H.C. Quake, S.R., Anal Chem, 79, 7576-7579 (2007)
  • HLA human leukocyte antigen
  • the invention provides a universal approach to noninvasive detection of graft rejection in transplant patients which circumvents the potential problems of microchimerism from DNA from other foreign sources and is general for all organ recipients without consideration of gender.
  • a genetic fingerprint is generated for the donor organ. This approach allows for a reliable identification of sequences arising solely from the organ transplantation that can be made in a manner that is independent of the genders of donor and recipient.
  • both the donor and recipient will be genotyped prior to transplantation.
  • methods that can be used to genotyped the transplant donor and the transplant recipient include, but are not limited to, whole genome sequencing, exome sequencing, or polymorphisms arrays (e.g., SNP arrays).
  • a set of relevant and distinguishable markers between the two sources is established.
  • the set of markers comprises a set of polymorphic markers.
  • Polymorphic markers include single nucleotide polymorphisms (SNP's), restriction fragment length polymorphisms (RFLP's), short tandem repeats (STRs), variable number of tandem repeats (VNTR's), hypervariable regions, minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats, simple sequence repeats, and insertion elements such as Alu.
  • the set of markers comprises SNPs.
  • bodily fluid such as blood can be drawn from the patient and analyzed for markers.
  • bodily fluids include, but are not limited to, smears, sputum, biopsies, secretions, cerebrospinal fluid, bile, blood, lymph fluid, saliva, and urine.
  • Detection, identification and/or quantitation of the donor-specific markers can be performed using real-time PCR, chips (e.g., SNP chips), high-throughput shotgun sequencing of circulating nucleic acids (e.g. cell-free DNA), as well as other methods known in the art including the methods described herein.
  • the proportion of donor nucleic acids can be monitored over time and an increase in this proportion can be used to determine transplant status or outcome (e.g. transplant rejection).
  • detection, identification and/or quantitation of the donor-specific markers can be performed by mapping one or more nucleic acids (e.g., DNA) to the genome of the specie use to determine whether the one or more nucleic acids come from the transplant donor.
  • nucleic acids e.g., DNA
  • Polymorphic markers as described above can also be used where the transplant is a xenotransplant.
  • the transplant graft can be any solid organ or skin transplant.
  • organ transplants that can be analyzed by the methods described herein include but are not limited to kidney transplant, pancreas transplant, liver transplant, heart transplant, lung transplant, intestine transplant, pancreas after kidney transplant, and simultaneous pancreas-kidney transplant.
  • target nucleic acids are from a sample obtained from a subject that has received a transplant.
  • a subject can be a human or a domesticated animal such as a cow, chicken, pig, horse, rabbit, dog, cat, or goat.
  • the cells used in the present invention are taken from a patient.
  • Samples derived from an animal can include, for example whole blood, sweat, tears, saliva, ear flow, sputum, lymph, bone marrow suspension, lymph, urine, saliva, semen, vaginal flow, cerebrospinal fluid, brain fluid, ascites, milk, secretions of the respiratory, intestinal or genitourinary tracts fluid, a lavage of a tissue or organ (e.g. lung) or tissue which has been removed from organs, such as breast, lung, intestine, skin, cervix, prostate, pancreas, heart, liver and stomach.
  • a tissue sample can comprise a region of functionally related cells or adjacent cells.
  • Such samples can comprise complex populations of cells, which can be assayed as a population, or separated into sub-populations.
  • Such cellular and acellular samples can be separated by centrifugation, elutriation, density gradient separation, apheresis, affinity selection, panning, FACS, centrifugation with Hypaque, etc.
  • a relatively homogeneous population of cells may be obtained.
  • a heterogeneous cell population can be used.
  • Cells can also be separated by using filters. For example, whole blood can also be applied to filters that are engineered to contain pore sizes that select for the desired cell type or class.
  • Cells can be filtered out of diluted, whole blood following the lysis of red blood cells by using filters with pore sizes between 5 to 10 ⁇ , as disclosed in U.S. Patent Application No. 09/790,673.
  • Other devices can separate cells from the bloodstream, see Demirci U, Toner M., Direct etch method for microfluidic channel and nanoheight post-fabrication by picoliter droplets, Applied Physics Letters 2006; 88 (5), 0531 17; and Irimia D, Geba D, Toner M., Universal microfluidic gradient generator, Analytical Chemistry 2006; 78: 3472-3477.
  • a sample Once a sample is obtained, it can be used directly, frozen, or maintained in appropriate culture medium for short periods of time. Methods to isolate one or more cells for use according to the methods of this invention are performed according to standard techniques and protocols well-established in the art.
  • a blood sample can be optionally pre-treated or processed prior to enrichment.
  • pre-treatment steps include the addition of a reagent such as a stabilizer, a preservative, a fixant, a lysing reagent, a diluent, an anti- apoptotic reagent, an anti-coagulation reagent, an anti-thrombotic reagent, magnetic property regulating reagent, a buffering reagent, an osmolality regulating reagent, a pH regulating reagent, and/or a cross-linking reagent.
  • a reagent such as a stabilizer, a preservative, a fixant, a lysing reagent, a diluent, an anti- apoptotic reagent, an anti-coagulation reagent, an anti-thrombotic reagent, magnetic property regulating reagent, a buffering reagent, an osmolality regulating reagent, a pH
  • a preservative such an anti-coagulation agent and/or a stabilizer can be added to the sample prior to enrichment. This allows for extended time for analysis/detection.
  • a sample such as a blood sample, can be analyzed under any of the methods and systems herein within 1 week, 6 days, 5 days, 4 days, 3 days, 2 days, 1 day, 12 hrs, 6 hrs, 3 hrs, 2 hrs, or 1 hr from the time the sample is obtained.
  • a blood sample can be combined with an agent that selectively lyses one or more cells or components in a blood sample.
  • an agent that selectively lyses one or more cells or components in a blood sample For example platelets and/or enucleated red blood cells are selectively lysed to generate a sample enriched in nucleated cells.
  • the cells of interest can subsequently be separated from the sample using methods known in the art.
  • the amount can vary depending upon subject size and the condition being screened. In some embodiments, up to 50, 40, 30, 20, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 ml. of a sample is obtained. In some embodiments, 1-50, 2-40, 3-30, or 4-20 ml. of sample is obtained. In some embodiments, more than 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 100 ml_ of a sample is obtained.
  • Nucleic acids from samples that can be analyzed by the methods herein include: double-stranded DNA, single-stranded DNA, single-stranded DNA hairpins, DNA RNA hybrids, RNA (e.g. mRNA or miRNA) and RNA hairpins.
  • RNA e.g. mRNA or miRNA
  • Examples of genetic analyses that can be performed on nucleic acids include e.g., sequencing, SNP detection, STR detection, RNA expression analysis, and gene expression.
  • less than 1 pg, 5pg, 10 pg, 20 pg, 30 pg, 40 pg, 50 pg, 100 pg, 200 pg, 500 pg, 1 ng , 5ng, 10 ng, 20 ng, 30 ng, 40 ng, 50 ng, 100 ng, 200 ng, 500 ng, 1 ug, 5ug, 10 ug, 20 ug, 30 ug, 40 ug, 50 ug, 100 ug, 200 ug, 500 ug or 1 mg of nucleic acids are obtained from the sample for further genetic analysis.
  • nucleic acids are obtained from the sample for further genetic analysis.
  • the methods described herein are used to detect and/or quantified a target nucleic acid molecule. In some embodiments, the methods described herein are used to detect and/or quantified multiple target nucleic acid molecules.
  • the methods described herein can analyzed at least 1 ; 2; 3; 4; 5; 10, 20; 50; 100; 200; 500; 1 ,000; 2,000; 5,000; 10,000, 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1 ,000,000; 2,000,000 or 3,000,000 different target nucleic acids.
  • the methods described herein are used to distinguish between target nucleic acids that differ from another nucleic acid by 1 nt. In some embodiments, the methods described herein are used to distinguish between target nucleic acids that differ from another nucleic acid by 1 nt or more than 1 , 2, 3, 5, 10, 15, 20, 21 , 22, 24, 25, 30 nt.
  • the methods described herein are used to detect and/or quantify genomic DNA regions.
  • the methods described herein can discriminate and quantitate genomic DNA regions.
  • the methods described herein can discriminate and quantitate at least 1 ; 2; 3; 4; 5; 10, 20; 50; 100; 200; 500; 1 ,000; 2,000; 5,000; 10,000, 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1 ,000,000; 2,000,000 or 3,000,000 different genomic DNA regions.
  • the methods described herein can discriminate and quantitate genomic DNA regions varying by 1 nt or more than 1 , 2, 3, 5, 10, 15, 20, 21 , 22, 24, 25, 30 nt.
  • the methods described herein are used to detect and/or quantify genomic DNA regions such as a region containing a DNA polymorphism.
  • a polymorphism refers to the occurrence of two or more genetically determined alternative sequences or alleles in a population.
  • a polymorphic marker or site is the locus at which divergence occurs. Preferred markers have at least two alleles, each occurring at a frequency of preferably greater than 1 %, and more preferably greater than 10% or 20% of a selected population.
  • a polymorphism may comprise one or more base changes, an insertion, a repeat, or a deletion.
  • a polymorphic locus may be as small as one base pair.
  • Polymorphic markers include single nucleotide polymorphisms (SNP's), restriction fragment length polymorphisms (RFLP's), short tandem repeats (STRs), variable number of tandem repeats (VNTR's), hypervariable regions, minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats, simple sequence repeats, and insertion elements such as Alu.
  • SNP's single nucleotide polymorphisms
  • RFLP's restriction fragment length polymorphisms
  • STRs short tandem repeats
  • VNTR's variable number of tandem repeats
  • hypervariable regions minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats, simple sequence repeats, and insertion elements such as Alu.
  • a polymorphism between two nucleic acids can occur naturally, or be caused by exposure to or contact with chemicals, enzymes
  • the methods described herein can discriminate and quantitate a DNA region containing a DNA polymorphism.
  • the methods described herein can discriminate and quantitate of at least 1 ; 2; 3; 4; 5; 10, 20; 50; 100; 200; 500; 1 ,000; 2,000; 5,000; 10,000, 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1 ,000,000; 2,000,000 or 3,000,000 DNA polymorphism.
  • the methods described herein can discriminate and quantitate at least 1 ; 2; 3; 4; 5; 10; 20; 50; 100; 200; 500; 1 ,000; 2,000; 5,000; 10,000; 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1 ,000,000; 2,000,000 or 3,000,000 different polymorphic markers.
  • the methods described herein can discriminate and quantitate at least 1 ; 2; 3; 4; 5; 10; 20; 50; 100; 200; 500; 1 ,000; 2,000; 5,000; 10,000; 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1 ,000,000; 2,000,000 or 3,000,000 different SNPs.
  • the methods described herein are used to detect and/or quantify gene expression. In some embodiments, the methods described herein provide high discriminative and quantitative analysis of multiples genes. The methods described herein can discriminate and quantitate the expression of at least 1 , 2, 3, 4, 5, 10, 20, 50, 100, 200, 500, 1 ,000, 2,000, 5,000, 10,000, 20,000, 50,000, 100,000, different target nucleic acids.
  • the methods described herein are used to detect and/or quantify gene expression of genes with similar sequences.
  • the methods described herein can discriminate and quantitate the expression of genes varying by 1 nt or more than 1 , 2, 3, 4, 5, 10, 12, 15, 20, 21 , 22, 24, 25, 30 nt.
  • the methods described herein are used to detect and/or quantify genomic DNA regions by mapping the region to the genome of a species in the case where the transplant donor and the transplant recipient are not from the same species (e.g., xenotransplants). In some embodiments, the methods described herein can discriminate and quantitate a DNA region from a species.
  • the methods described herein can discriminate and quantitate of at least 1 ; 2; 3; 4; 5; 10, 20; 50; 100; 200; 500; 1 ,000; 2,000; 5,000; 10,000, 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1 ,000,000; 2,000,000 or 3,000,000 DNA regions from a species.
  • the methods described herein are used for diagnosing or predicting transplant status or outcome (e.g. transplant rejection). In some embodiments, the methods described herein are used to detect and/or quantify target nucleic acids to determine whether a patient or subject is displaying transplant tolerance. In some embodiments, the methods described herein are used to detect and/or quantify target nucleic acids for diagnosis or prediction of transplant rejection. In some embodiments, the methods described herein are used to detect and/or quantify target nucleic acids for determining an immunosuppressive regimen for a subject who has received a transplant, e.g., an allograft.
  • a transplant status or outcome e.g. transplant rejection
  • the methods described herein are used to detect and/or quantify target nucleic acids to determine whether a patient or subject is displaying transplant tolerance. In some embodiments, the methods described herein are used to detect and/or quantify target nucleic acids for diagnosis or prediction of transplant rejection. In some embodiments, the methods described herein are used to detect and/or quantify target nucleic acids for determining
  • the methods described herein are used to detect and/or quantify target nucleic acids to predict transplant survival in a subject that have received a transplant.
  • the invention provides methods of diagnosing or predicting whether a transplant in a transplant patient or subject will survive or be lost.
  • the methods described herein are used to detect and/or quantify target nucleic acids to diagnose or predict the presence of long-term graft survival.
  • the methods described herein are used to detect and/or quantify target nucleic acids for diagnosis or prediction of non-rejection based transplant injury. Examples of non-rejection based graft injury include, but are not limited to, ischemic injury, virus infection, perioperative ischemia, reperfusion injury, hypertension, physiological stress, injuries due to reactive oxygen species and injuries caused by pharmaceutical agents.
  • the term "diagnose” or “diagnosis” of a transplant status or outcome includes predicting or diagnosing the transplant status or outcome, determining predisposition to a transplant status or outcome, monitoring treatment of transplant patient, diagnosing a therapeutic response of transplant patient, and prognosis of transplant status or outcome, transplant progression, and response to particular treatment.
  • the methods, devices, compositions and kits are used to establish a genotype for both the donor and the recipient before transplantation to enable the detection of donor-specific nucleic acids such as DNA or RNA in bodily fluids such as blood or urine from the organ recipient after transplantation.
  • This approach allows for a reliable identification of sequences arising solely from the organ transplantation that can be made in a manner that is independent of the genders of donor and recipient.
  • a genetic fingerprint is generated for the donor organ. Both the donor and recipient will be genotyped prior to transplantation. Genotyping of transplant donors and transplant recipients establishes a profile, using distinguishable markers, for detecting donor nucleic acids (e.g. circulating cell-free nucleic acid or nucleic acids from circulating donor cells). In some embodiments, for xenotransplants, nucleic acids from the donors can be mapped to the genome of the donor species.
  • donor nucleic acids e.g. circulating cell-free nucleic acid or nucleic acids from circulating donor cells.
  • nucleic acids from the donors can be mapped to the genome of the donor species.
  • samples as described above can be drawn from the patient and analyzed for markers.
  • the proportion of donor nucleic acids can be monitored over time and an increase in this proportion can be used to determine transplant status or outcome (e.g. transplant rejection).
  • genotyping comprises detection and quantitation of nucleic acids from circulating transplant donor cells or circulating cell-free nucleic acids.
  • nucleic acids include, but are not limited to double-stranded DNA, single-stranded DNA, single-stranded DNA hairpins, DNA RNA hybrids, RNA (e.g. mRNA or miRNA) and RNA hairpins.
  • the nucleic acid is DNA.
  • the nucleic acid is RNA.
  • cell-free RNA is also present in human plasma (Tong, Y.K.
  • genotyping comprises detection and quantitation of polymorphic markers.
  • polymorphic markers include single nucleotide polymorphisms (SNP's), restriction fragment length polymorphisms (RFLP's), variable number of tandem repeats (VNTR's), short tandem repeats (STRs), hypervariable regions, minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats, simple sequence repeats, and insertion elements such as Alu.
  • SNP's single nucleotide polymorphisms
  • RFLP's restriction fragment length polymorphisms
  • VNTR's variable number of tandem repeats
  • STRs short tandem repeats
  • hypervariable regions minisatellites, dinucleotide repeats, trinucleotide repeats, tetranucleotide repeats, simple sequence repeats, and insertion elements such as Alu.
  • genotyping comprises detection and quantitation of STRs.
  • genotyping comprises detection and quantitation of VNTRs
  • genotyping comprises detection and quantitation of SNPs.
  • any donor and recipient will vary at roughly three million SNP positions if fully genotyped.
  • Usable SNPs must be homozygous for the recipient and ideally homozygous for the donor as well. While the majority of these positions will contain SNPs that are heterozygous for either the donor or the recipient, over 10% (or hundreds of thousands) will be homozygous for both donor and recipient meaning a direct read of that SNP position can distinguish donor DNA from recipient DNA.
  • genotyping a transplant donor and transplant recipient using existing genotyping platforms know in the art including the one described herein, one could identify approximately 1.2 million total variations between a transplant donor and transplant recipient.
  • Usable SNPs may comprise approximately 500,000 heterozygous donor SNPs and approximately 160,000 homozygous donor SNPs.
  • Companies such as Applied Biosystems, Inc.
  • TaqMan probe sets for SNP genotyping that can in principle target any desired SNP position for a PCR-based assay (Livak, K. L, Marmaro, J., Todd, J. A., Nature Genetics, 9, 341-342 (1995); De La Vefa, F. M., Lazaruk, K. D., Rhodes, M. D., Wenz, M. H., Mutation Research, 573, 1 1 1-135 (2005)).
  • a usable subset of existing or custom probes can be selected to serve as the probe set for any donor/recipient pair.
  • digital PCR or real-time PCR performed on the nucleic acids recovered from plasma or other biological samples will directly quantitate the percentage of donor-specific species seen in the sample.
  • sequencing performed on the nucleic acid recovered from plasma or other biological samples will directly quantitate the percentage of donor-specific species seen in the sample.
  • arrays can be used on the nucleic acids recovered from plasma or other biological samples to directly quantitate the percentage of donor-specific species seen in the sample.
  • Pre-amplification can be performed using any suitable method known in the art such as multiple displacement amplification (MDA) (Gonzalez et al. Envircon Microbiol; 7(7); 1024-8 (2005)) or amplification with outer primers in a nested PCR approach.
  • MDA multiple displacement amplification
  • donor nucleic acids This permits detection and analysis of donor nucleic acids even if the total amount of donor nucleic acid in the sample (e.g. blood from transplant patient) is only up to 1 ⁇ g, 500 ng, 200 ng, 100 ng, 50 ng, 40 ng, 30 ng, 20 ng, 10 ng, 5ng, 1 ng , 500 pg, 200 pg, 100 pg, 50 pg, 40 pg, 30 pg, 20 p, 10 pg, 5pg, or 1 pg or between 1 5 ⁇ g, 5 10 Mg, or 10 50 pg.
  • the sample e.g. blood from transplant patient
  • Genotyping donor and recipient nucleic acids, and/or detection, identification and/or quantitation of the donor-specific nucleic acids after transplantation can be performed by PCR.
  • PCR techniques that can be used to detect, identify and/or quantitate the donor-specific nucleic acids include, but are not limited, to quantitative PCR, quantitative fluorescent PCR (QF-PCR), multiplex fluorescent PCR (MF-PCR), real time PCR (RT-PCR), single cell PCR, restriction fragment length polymorphism PCR (PCR-RFLP), PCR-RFLP/RT-PCR-RFLP, hot start PCR, nested PCR, in situ polonony PCR, in situ rolling circle amplification (RCA), bridge PCR, picotiter PCR and emulsion PCR.
  • QF-PCR quantitative fluorescent PCR
  • MF-PCR multiplex fluorescent PCR
  • RT-PCR real time PCR
  • PCR-RFLP restriction fragment length polymorphism PCR
  • PCR-RFLP/RT-PCR-RFLP hot start
  • Suitable amplification methods include the ligase chain reaction (LCR), transcription amplification, self-sustained sequence replication, selective amplification of target polynucleotide sequences, consensus sequence primed polymerase chain reaction (CP-PCR), arbitrarily primed polymerase chain reaction (AP-PCR), degenerate oligonucleotide-primed PCR (DOP-PCR) and nucleic acid based sequence amplification (NABSA).
  • Other amplification methods that may be used to amplify specific polymorphic loci include those described in, U.S. Pat. Nos. 5,242,794, 5,494,810, 4,988,617 and 6,582,938.
  • Detection, identification and/or quantitation of the donor-specific nucleic acids is performed by realtime PCR.
  • digital PCR or real time PCR to quantitate the presence of specific polymorphisms that have already been identified in the initial genotyping step pre- transplantation.
  • digital PCR is a much more accurate and reliable method to quantitate nucleic acid species including rare nucleic acid species, and does not require a specific gender relationship between donor and recipient. (Warren, L, Bryder, D., Weissman, I.L., Quake, S.R., Proc Natl Acad Sci, 103, 17807-17812 (2006)).
  • digital PCR or real-time PCR assays can be used to quantitate the fraction of donor DNA in a transplant patient using probes targeted to several SNPs.
  • Genotyping donor and recipient nucleic acids, and/or detection, identification and/or quantitation of the donor-specific nucleic acids after transplantation can be performed by sequencing such as whole genome sequencing or exome sequencing. Sequencing can be accomplished through classic Sanger sequencing methods which are well known in the art. Sequence can also be accomplished using high-throughput systems some of which allow detection of a sequenced nucleotide immediately after or upon its incorporation into a growing strand, i.e., detection of sequence in red time or substantially real time.
  • high throughput sequencing generates at least 1 ,000, at least 5,000, at least 10,000, at least 20,000, at least 30,000, at least 40,000, at least 50,000, at least 100,000 or at least 500,000 sequence reads per hour; with each read being at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 120 or at least 150 bases per read.
  • Sequencing can be performed using nucleic acids described herein such as genomic DNA, cDNA derived from RNA transcripts or RNA as a template.
  • high-throughput sequencing involves the use of technology available by Helicos Biosciences Corporation (Cambridge, Massachusetts) such as the Single Molecule Sequencing by Synthesis (SMSS) method.
  • SMSS is unique because it allows for sequencing the entire human genome with no pre amplification step needed. Thus, distortion and nonlinearity in the measurement of nucleic acids are reduced.
  • This sequencing method also allows for detection of a SNP nucleotide in a sequence in substantially real time or real time.
  • SMSS is powerful because, like the MIP technology, it does not require a pre amplification step prior to hybridization. In fact, SMSS does not require any amplification. SMSS is described in part in US Publication Application Nos. 2006002471 I; 20060024678; 20060012793; 20060012784; and 20050100932.
  • high-throughput sequencing involves the use of technology available by 454 Lifesciences, Inc. (Branford, Connecticut) such as the Pico Titer Plate device which includes a fiber optic plate that transmits chemiluninescent signal generated by the sequencing reaction to be recorded by a CCD camera in the instrument.
  • This use of fiber optics allows for the detection of a minimum of 20 million base pairs in 4.5 hours.
  • high-throughput sequencing is performed using Clonal Single Molecule Array (Solexa, Inc.) or sequencing-by-synthesis (SBS) utilizing reversible terminator chemistry.
  • Solexa, Inc. Clonal Single Molecule Array
  • SBS sequencing-by-synthesis
  • anyDot.chips Genevoxx, Germany
  • biological processes e.g., miRNA expression or allele variability (SNP detection).
  • SNP detection e.g., miRNA expression or allele variability
  • the AnyDot-chips allow for 10x - 50x enhancement of nucleotide fluorescence signal detection.
  • AnyDot.chips and methods for using them are described in part in International Publication Application Nos. WO 02088382, WO 03020968, WO 0303 1947, WO 2005044836, PCTEP 05105657, PCMEP 05105655; and German Patent Application Nos.
  • Sequence can then be deduced by identifying which base is being incorporated into the growing complementary strand of the target nucleic acid by the catalytic activity of the nucleic acid polymerizing enzyme at each step in the sequence of base additions.
  • a polymerase on the target nucleic acid molecule complex is provided in a position suitable lo move along the target nucleic acid molecule and extend the oligonucleotide primer at an active site.
  • a plurality of labeled types of nucleotide analogs are provided proximate to the active site, with each distinguishably type of nucleotide analog being complementary to a different nucleotide in the target nucleic acid sequence.
  • the growing nucleic acid strand is extended by using the polymerase to add a nucleotide analog to the nucleic acid strand at the active site, where the nucleotide analog being added is complementary to the nucleotide of the target nucleic acid at the active site.
  • the nucleotide analog added to the oligonucleotide primer as a result of the polymerizing step is identified.
  • the steps of providing labeled nucleotide analogs, polymerizing the growing nucleic acid strand, and identifying the added nucleotide analog are repeated so that the nucleic acid strand is further extended and the sequence of the target nucleic acid is determined. [0096] In some embodiments, shotgun sequencing is performed.
  • DNA is broken up randomly into numerous small segments, which are sequenced using the chain termination method to obtain reads. Multiple overlapping reads for the target DNA are obtained by performing several rounds of this fragmentation and sequencing. Computer programs then use the overlapping ends of different reads to assemble them into a continuous sequence
  • the invention provides methods for detection and quantitation of SNPs using sequencing.
  • depth of sequencing a frequent estimate for the variation between individuals is that about one base per thousand differs.
  • sequencers such as the lllumina Genome Analyzer have read lengths exceeding 36 base pairs. Without intending to be limited to any theory or specific embodiment, this means that roughly one in 30 molecules analyzed will have a potential SNP.
  • the sequencing error rate also affects the sensitivity of this technique. For an average error rate of ⁇ , the chance of a single SNP being accidentally identified as of donor origin as a result of a mis-read is roughly ⁇ /3. For each individual read, this establishes a lower limit of sensitivity of one's ability to determine whether the read is due to donor or recipient. Typical sequencing error rates for base substitutions vary between platforms, but are between 0.5-1.5%. This places a potential limit on sensitivity of 0.16 to 0.50%.
  • the informatics analysis independently measures the frequency of incorrect assignments for a given sample by examining SNP positions for which both the donor and recipient are homozygous and carry the same allele. The frequency of erroneous calls is then proportional to the frequency at which a base other than the donor and recipient allele is measured at these homozygous positions. The net effect of these assignment errors (recipient sequences assigned to donor or donor sequences assigned to the recipient) is an overestimate of the donor fraction.
  • the scaling with a > 1 is a consequence of (i) the dependence of the probability of an erroneous assignment on the frequency of occurrence of the allele in the population, as expected for genotyping assays, and (ii) differences in the distribution of allele frequencies for the set of SNPs used to evaluate the donor fraction and the matched error rate, respectively: SNPs that were used to estimate the donor fraction had a lower average recipient allele frequency than SNP markers that were used to estimate the error rate.
  • the expected scaling factor may be calculated from the from the measured distributions of allele frequencies for the different set of SNPs and the measured allele frequency dependence of the error rate.
  • low quality SNPs may be discarded, for example only retaining SNPs with high GenCall, for example greater than about 7, and Cluster Separation, for example equal to one, scores.
  • Base calls with a reported sequencing error rates higher than 2 ⁇ 10 3 may be excluded.
  • a Receiver-Operating Characteristics (ROC) analysis is performed on the performance of cfdDNA as marker of acute cellular rejection.
  • ROC Receiver-Operating Characteristics
  • an AUC is at least 0.7, at least 0.75, at least 0.8, at least 0.85, at least 0.9, at least 0.94 or more.
  • Genotyping donor and recipient nucleic acids, and/or detection, identification and/or quantitation of the donor-specific nucleic acids after transplantation can be performed using arrays (e.g. SNPs arrays). Results can be visualized using a scanner that enables the viewing of intensity of data collected and software to detect and quantify nucleic acid.
  • arrays e.g. SNPs arrays.
  • the transplant donor and/or recipient nucleic acids can be labeled and hybridized with a DNA microarray (e.g., 100K Set Array or other array). Results can be visualized using a scanner that enables the viewing of intensity of data collected and software "calls" the SNP present at each of the positions analyzed.
  • a DNA microarray e.g. 100K Set Array or other array.
  • genotyping microarrays that are used to detect SNPs can be used in combination with molecular inversion probes (MIPS) as described in Hardenbol et al., Genome Res. 15(2) :269-275,2005, Hardenbol, P. et al. Nature Biotechnology 2 1 (6), 673-8,2003; Faham M, et al. Hum Mol Genet. Aug 1 ; 10(16): 1657- 64,200 1 : Maneesh Jain, Ph.D., et al. Genetic Engineering News V24: No. 18, 2004; and Fakhrai-Rad H, el al. Genome Res. Jul; 14(7):1404-12, 2004; and in U.S.
  • MIPS molecular inversion probes
  • MIP technology involves the use enzymological reactions that can score up to 10,000: 20,000, 50,000; 100,000; 200,000; 500,000; 1 ,000,000; 2,000,000 or 5,000,000 SNPs (target nucleic acids) in a single assay.
  • SNPs target nucleic acids
  • the enzymological reactions are insensitive to crossreactivity among multiple probe molecules and there is no need for pre-amplification prior to hybridization of the probe with the genomic DNA.
  • the target nucleic acid(s) or SNPs can be obtained from a single cell.
  • nucleic acids are quantified.
  • Methods for quantifying nucleic acids include, but are not limited to, gas chromatography, supercritical fluid chromatography, liquid chromatography (including partition chromatography, adsorption chromatography, ion exchange chromatography, size exclusion chromatography, thin-layer chromatography, and affinity chromatography), electrophoresis (including capillary electrophoresis, capillary zone electrophoresis, capillary isoelectric focusing, capillary electrochromatography, micellar electrokinetic capillary chromatography, isotachophoresis, transient isotachophoresis and capillary gel electrophoresis), comparative genomic hybridization (CGH), microarrays, bead arrays, and high-throughput genotyping such as with the use of molecular inversion probe (MIP).
  • MIP molecular inversion probe
  • Another method contemplated by the present invention to detect and/or quantify target nucleic acids involves the use of nanoreporters as described in US patent 7,473,767 entitled “Methods for detection and quantification of analytes in complex mixtures", US patent publication no. 2007/0166708 entitled “Methods for detection and quantification of analytes in complex mixtures”, US application number 1 1/645,270 entitled “Compositions comprising oriented, immobilized macromolecules and methods for their preparation", PCT application no US06/049274 entitled “Nanoreporters and methods of manufacturing and use thereof", [00108] Quantification of target nucleic acid can be used to determine the percentage of donor nucleic acids such as DNA.
  • Detection and/or quantification of target nucleic acids can be done using fluorescent dyes known in the art.
  • Fluorescent dyes may typically be divided into families, such as fluorescein and its derivatives; rhodamine and its derivatives; cyanine and its derivatives; coumarin and its derivatives; Cascade BlueTM and its derivatives; Lucifer Yellow and its derivatives; BODIPY and its derivatives; and the like.
  • fluorophores include indocarbocyanine (C3), indodicarbocyanine (C5), Cy3, Cy3.5, Cy5, Cy5.5, Cy7, Texas Red, Pacific Blue, Oregon Green 488, Alexa fluor ® -355, Alexa Fluor 488, Alexa Fluor 532, Alexa Fluor 546, Alexa Fluor-555, Alexa Fluor 568, Alexa Fluor 594, Alexa Fluor 647, Alexa Fluor 660, Alexa Fluor 680, JOE, Lissamine, Rhodamine Green, BODIPY, fluorescein isothiocyanate (FITC), carboxy-fluorescein (FAM), phycoerythrin, rhodamine, dichlororhodamine (dRhodamineTM ), carboxy tetramethylrhodamine (TAMRATM ), carboxy- X-rhodamine (ROX.TM.), LIZTM, VICTM., NEDTM, PET
  • a branched-DNA (bDNA) approach is used to increase the detection sensitivity.
  • bDNA approach is applied to an array detection assay.
  • the array detection assay can be any array assay known in the art, including the array assays described herein.
  • bDNA approach amplifies the signals through a branched DNA that are attached by tens or hundreds of alkaline phosphatase molecules. Thus, the signals are significantly amplified while the fidelity of the original nucleic acid target abundance is maintained.
  • the invention provides methods for the diagnosis or prediction of transplant status or outcome in a subject who has received a transplant.
  • the transplant status or outcome may comprise rejection, tolerance, non-rejection based transplant injury, transplant function, transplant survival, chronic transplant injury, or titer pharmacological immunosuppression.
  • non-rejection based allograft injury include, but are not limited to, ischemic injury, virus infection, peri-operative ischemia, reperfusion injury, hypertension, physiological stress, injuries due to reactive oxygen species and injuries caused by pharmaceutical agents.
  • the transplant status or outcome may comprise vascular complications or neoplastic involvement of the transplanted organ.
  • the invention provides methods of diagnosing or predicting transplant status or outcome comprising the steps of: (i) providing a sample from a subject who has received a transplant from a donor; (ii) determining the presence or absence of one or more nucleic acids from the donor transplant, wherein the one or more nucleic acids from the donor are identified based on a predetermined marker profile; and (iii) diagnosing or predicting transplant status or outcome based on the presence or absence of the one or more nucleic acids from said donor.
  • the methods of the invention are used to establish a genotype for both the donor and the recipient before transplantation.
  • the genotyping of both the donor and the recipient before transplantation enables the detection of donor-specific nucleic acids such as DNA or RNA in bodily fluids as described herein (e.g., blood or urine) from the organ recipient after transplantation.
  • a marker profile for the donor is determined based on the genotyping of the transplant donor.
  • a marker profile is determined for the transplant recipient based on the genotyping of the transplant recipient.
  • a marker profile is established by selecting markers that are distinguishable between the transplant donor and the subject receiving the transplant. This approach allows for a reliable identification of nucleic acids arising solely from the organ transplantation that can be made in a manner that is independent of the genders of donor and recipient.
  • Genotyping of the transplant donor and/or the transplant recipient may be performed by any suitable method known in the art including those described herein such as sequencing, nucleic acid array or PCR. In some embodiments, genotyping of the transplant donor and/or the transplant recipient is performed by shotgun sequencing. In some embodiments, genotyping of the transplant donor and/or the transplant recipient is performed using a DNA array. In some embodiments, genotyping of the transplant donor and/or the transplant recipient is performed using a polymorphism array such as a SNP array.
  • the marker profile is a polymorphic marker profile.
  • Polymorphic marker profile may comprise one or more single nucleotide polymorphisms (SNP's), one or more restriction fragment length polymorphisms (RFLP's), one or more short tandem repeats (STRs), one or more variable number of tandem repeats (VNTR's), one or more hypervariable regions, one or more minisatellites, one or more dinucleotide repeats, one or more trinucleotide repeats, one or more tetranucleotide repeats, one or more simple sequence repeats, or one or more insertion elements.
  • SNP's single nucleotide polymorphisms
  • RFLP's restriction fragment length polymorphisms
  • STRs short tandem repeats
  • VNTR's variable number of tandem repeats
  • hypervariable regions one or more minisatellites, one or more dinucleotide repeats, one or more trinucleotide repeats, one or more tetranucleotide repeats, one
  • the marker profile comprises at least 1 ; 2; 3; 4; 5; 10; 20; 50; 100; 200; 500; 1 ,000; 2,000; 5,000; 10,000; 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1 ,000,000; 2,000,000 or 3,000,000 different polymorphic markers.
  • the polymorphic marker profile comprises one or more SNPs.
  • the marker profile comprises at least 1 ; 2; 3; 4; 5; 10; 20; 50; 100; 200; 500; 1 ,000; 2,000; 5,000; 10,000; 20,000; 50,000; 100,000; 200,000; 300,000; 400,000; 500,000; 600,000; 700,000; 800,000; 900,000; 1 ,000,000; 2,000,000 or 3,000,000 different SNPs.
  • samples as described above can be drawn from the patient and analyzed for the presence or absence of one or more nucleic acids from the transplant donor.
  • the sample is blood, plasma, serum or urine.
  • the proportion and/or amount of donor nucleic acids can be monitored over time and an increase in this proportion can be used to determine transplant status or outcome (e.g. transplant rejection).
  • the presence or absence of one or more nucleic acids from the transplant donor in the transplant recipient may be determined by any suitable method known in the art including those described herein such as sequencing, nucleic acid arrays or PCR. In some embodiments, the presence or absence of one or more nucleic acids from the transplant donor in the transplant recipient is determined by shotgun sequencing. In some embodiments, the presence or absence of one or more nucleic acids from the transplant donor in the transplant recipient is determined using a DNA array. In some embodiments, the presence or absence of one or more nucleic acids from the transplant donor in the transplant recipient is determined using a polymorphism array such as a SNP array.
  • detection, identification and/or quantitation of the donor-specific markers can be performed by mapping one or more nucleic acids (e.g., DNA) to the genome of the specie use to determine whether the one or more nucleic acids come from the transplant donor.
  • nucleic acids e.g., DNA
  • Polymorphic markers as described above can also be used where the transplant is a xenotransplant.
  • the presence or absence of circulating DNA or RNA from a transplant donor in a transplant recipient is used to determine the transplant status or outcome.
  • the DNA can be double-stranded DNA, single-stranded DNA, single-stranded DNA hairpins, or cDNA.
  • the RNA can be single stranded RNA or RNA hairpins.
  • the presence or absence of circulating DNA/RNA hybrids from a transplant donor in a transplant recipient is used to determine the transplant status or outcome.
  • the presence or absence of circulating mRNA from a transplant donor in a transplant recipient is used to determine the transplant status or outcome.
  • the presence or absence of circulating DNA from a transplant donor in a transplant recipient is used to determine the transplant status or outcome.
  • cDNA is used to determine the transplant status or outcome.
  • the DNA or RNA can be obtained from circulating donor cells.
  • the DNA or RNA can be circulating cell-free DNA or circulating cell-free RNA
  • the transplant graft maybe any solid organ and skin transplant.
  • transplants whose transplant status or outcome could be determined by the methods described herein, include but are not limited to, kidney transplant, heart transplant, liver transplant, pancreas transplant, lung transplant, intestine transplant and skin transplant.
  • the invention provides methods of determining whether a patient or subject is displaying transplant tolerance. In some embodiments the invention provides methods for diagnosis or prediction of transplant rejection. The term "transplant rejection" encompasses both acute and chronic transplant rejection. In some embodiments, the invention further includes methods for determining an immunosuppressive regimen for a subject who has received a transplant, e.g., an allograft. In some embodiments, the invention further includes methods for determining the effectiveness of an immunosuppressive regimen for a subject who has received a transplant. Certain embodiments of the invention provide methods of predicting transplant survival in a subject that has received a transplant. The invention provides methods of diagnosing or predicting whether a transplant in a transplant patient or subject will survive or be lost.
  • the invention provides methods of diagnosing or predicting the presence of long-term graft survival.
  • the invention provides methods for diagnosis or prediction of non-rejection based transplant injury.
  • non-rejection based transplant injury include, but are not limited to, ischemic injury, virus infection, perioperative ischemia, reperfusion injury, hypertension, physiological stress, injuries due to reactive oxygen species and injuries caused by pharmaceutical agents.
  • the invention provides methods for diagnosis or prediction of vascular complications or neoplastic involvement of the transplanted organ.
  • the amount of one or more nucleic acids from the transplant donor in a sample from the transplant recipient is used to determine the transplant status or outcome.
  • the methods of the invention further comprise quantitating the one or more nucleic acids from the transplant donor.
  • the amount of one or more nucleic acids from the donor sample is determined as a percentage of total the nucleic acids in the sample.
  • the amount of one or more nucleic acids from the donor sample is determined as a ratio of the total nucleic acids in the sample.
  • the amount of one or more nucleic acids from the donor sample is determined as a ratio or percentage compared to one or more reference nucleic acids in the sample.
  • the amount of one or more nucleic acids from the transplant donor can be determined to be 10% of the total nucleic acids in the sample.
  • the amount of one or more nucleic acids from the transplant donor can be at a ratio of 1 :10 compared to total nucleic acids in the sample.
  • the amount of one or more nucleic acids from the transplant donor can be determined to be 10% or at a ratio of 1 :10 of a reference gene such a ⁇ -globin.
  • the amount of one or more nucleic acids from the transplant donor can be determined as a concentration.
  • the amount of one or more nucleic acids from the donor sample can be determined to be 1 ug/mL.
  • the amount of one or more nucleic acids from the transplant donor above a predetermined threshold value is indicative of a transplant status or outcome.
  • the normative values for clinically stable post-transplantation patients with no evidence of graft rejection or other pathologies can be determined.
  • An increase in the amount of one or more nucleic acids from the transplant donor above the normative values for clinically stable post-transplantation patients could indicate a change in transplant status or outcome such as transplant rejection or transplant injury.
  • an amount of one or more nucleic acids from the transplant donor below or at the normative values for clinically stable post-transplantation patients could indicate graft tolerance or graft survival.
  • different predetermined threshold values are indicative of different transplant outcomes or status. For example, as discussed above, an increase in the amount of one or more nucleic acids from the transplant donor above the normative values for clinically stable post-transplantation patients could indicate a change in transplant status or outcome such as transplant rejection or transplant injury. However, an increase in the amount of one or more nucleic acids from the transplant donor above the normative values for clinically stable post-transplantation patients but below a predetermined threshold level could indicate a less serious condition such as a viral infection rather than transplant rejection. An increase in the amount of one or more nucleic acids from the transplant donor above a higher threshold could indicate transplant rejection.
  • temporal differences in the amount of said one or more nucleic acids from the transplant donor are indicative of a transplant status or outcome.
  • a transplant patient can be monitored over time to determine the amount of one or more nucleic acids from the transplant donor.
  • a sustained increase in the amount one or more nucleic acids from the transplant donor might indicate a serious condition such as transplant rejection.
  • temporal differences in the amount of said one or more nucleic acids from the transplant donor can be used to monitor effectiveness of an immunosuppressant treatment or to select an immunosuppressant treatment.
  • the amount of one or more nucleic acids from the transplant donor can be determined before and after an immunosuppressant treatment.
  • a decrease in the one or more nucleic acids from the transplant donor after treatment may indicate that the treatment was successful in preventing transplant rejection.
  • the amount of one or more nucleic acids from the transplant donor can be used to choose between immunosuppressant treatments, for examples, immunosuppressant treatments of different strengths.
  • a higher amount in one or more nucleic acids from the transplant donor may indicate that there is a need of a very potent immunosuppressant, whereas a lower amount in one or more nucleic acids from the transplant donor may indicate that a less potent immunosuppressant may be used.
  • the invention provides methods that sensitive and specific.
  • the methods described herein for diagnosing or predicting transplant status or outcome have at least 56 %, 60%, 70%, 80%, 90%, 95% or 100% sensitivity.
  • the methods described herein have at least 56 % sensitivity.
  • the methods described herein have at least 78 % sensitivity.
  • the methods described herein have a specificity of about 70% to about 100%.
  • the methods described herein have a specificity of about 80% to about 100%.
  • the methods described herein have a specificity of about 90% to about 100%.
  • the methods described herein have a specificity of about 100%.
  • the donor nucleic acid is cell-free DNA or DNA isolated from circulating donor cells.
  • Donor nucleic acid can be identified by the methods described herein including the methods described in the Examples. After identifying these, then one could look at the donor nucleic acids and examine them for their correlation with transplant status and outcomes such as chronic graft injury, rejection, and tolerance. In some embodiments, the longitudinal change of donor nucleic acids is studied. If clinically significant, these levels could be followed to titer pharmacological immunosuppression, or could be studied as a target for depletion. [00131] Also provided are reagents and kits thereof for practicing one or more of the above- described methods. The subject reagents and kits thereof may vary greatly.
  • Reagents of interest include reagents specifically designed for use in production of the above-described: (i) genotyping of a transplant donor and a transplant recipient; (ii) identification of marker profiles; and (ii) detection and/or quantitation of one or more nucleic acids from a transplant donor in a sample obtained from a transplant recipient.
  • One type of such reagents are one or more probes or an array of probes to genotype and/or to detect and/or to quantitate one or more nucleic acids.
  • a variety of different array formats are known in the art, with a wide variety of different probe structures, substrate compositions and attachment technologies.
  • kits of the subject invention may include the above-described arrays. Such kits may additionally comprise one or more therapeutic agents.
  • the kit may further comprise a software package for data analysis, which may include reference profiles for comparison with the test profile.
  • kits may comprise reagents such as buffers, and H 2 0.
  • the kits may comprise reagents necessary to perform nucleic acid extraction and/or nucleic acid detection using the methods described herein such as PCR and sequencing.
  • kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider.
  • kits may also include instructions to access a database. Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
  • any of the methods above can be performed by a computer program product that comprises a computer executable logic that is recorded on a computer readable medium.
  • the computer program can execute some or all of the following functions: (i) controlling isolation of nucleic acids from a sample, (ii) pre-amplifying nucleic acids from the sample, (iii) amplifying, sequencing or arraying specific polymorphic regions in the sample, (iv) identifying and quantifying a marker profile in the sample, (v) comparing data on marker profile detected from the sample with a predetermined threshold, (vi) determining a transplant status or outcome , (vi) declaring normal or abnormal transplant status or outcome.
  • the computer executable logic can analyze data on the detection and quantity of polymorphism(s) (e.g. SNPs).
  • the computer executable logic can work in any computer that may be any of a variety of types of general-purpose computers such as a personal computer, network server, workstation, or other computer platform now or later developed.
  • a computer program product is described comprising a computer usable medium having the computer executable logic (computer software program, including program code) stored therein.
  • the computer executable logic can be executed by a processor, causing the processor to perform functions described herein.
  • some functions are implemented primarily in hardware using, for example, a hardware state machine. Implementation of the hardware state machine so as to perform the functions described herein will be apparent to those skilled in the relevant arts.
  • the program can provide a method of evaluating a transplant status or outcome in a transplant recipient by accessing data that reflects the genotyping of the transplant donor and the transplant patient, and/or the presence or absence of one or more nucleic acids from the transplant donor in the circulation of the transplant patient post-transplantation.
  • the computer executing the computer logic of the invention may also include a digital input device such as a scanner.
  • the digital input device can provide information on a nucleic acid, e.g., polymorphism levels/quantity.
  • a scanner of this invention can provide an image of the polymorphism (e.g., SNPs) according to method herein.
  • a scanner can provide an image by detecting fluorescent, radioactive, or other emission; by detecting transmitted, reflected, or scattered radiation; by detecting electromagnetic properties or other characteristics; or by other techniques.
  • the data detected is typically stored in a memory device in the form of a data file.
  • a scanner may identify one or more labeled targets.
  • a first DNA polymorphism may be labeled with a first dye that fluoresces at a particular characteristic frequency, or narrow band of frequencies, in response to an excitation source of a particular frequency.
  • a second DNA polymorphism may be labeled with a second dye that fluoresces at a different characteristic frequency.
  • the excitation sources for the second dye may, but need not, have a different excitation frequency than the source that excites the first dye, e.g., the excitation sources could be the same, or different, lasers.
  • the invention provides a computer readable medium comprising a set of instructions recorded thereon to cause a computer to perform the steps of (i) receiving data from one or more nucleic acids detected in a sample from a subject who has received transplant from a donor, wherein said one or more nucleic acids are nucleic acids from said donor transplant, and wherein said one or more nucleic acids from said donor are identified based on a predetermined marker profile; and (ii) diagnosing or predicting transplant status or outcome based on the presence or absence of the one or more nucleic acids.
  • GTD genome transplant dynamics
  • Figure 2A illustrates the working principle of the assay used to quantify the fraction of donor-derived cell-free DNA. Circulating cell-free DNA was purified from the plasma samples collected post-transplant and sequenced (mean depth 1.2 Gbp, 24 million 50 bp reads per sample, Fig. 2B). The SNP-genotyping information obtained pre-transplant was used to discriminate donor- and recipient-derived sequences. SNPs were selected from single-base alleles that were distinct between donor and recipient and homozygous within each individual (e.g. marker n Fig. 2A, for details see methods). On average, 35,673 informative SNP markers were available per genotype pair.
  • Figure 2C shows a histogram of the number of reads that overlapped with informative SNPs across all samples (mean 13,042 SNPs).
  • the donor fraction was calculated as N D/N, where N is the total number of assignments made in sequencing and N D is the number of donor- derived sequences.
  • a histogram of the number of donor assignments is shown in Figure 2D.
  • Rate of incorrect sequence assignments Errors introduced in sequencing and genotyping can potentially give rise to incorrect donor or recipient assignments.
  • the frequency of erroneous calls is then proportional to the frequency at which a base other than the donor and recipient allele is measured at these homozygous positions (see Materials and Methods).
  • Fig. 3A shows a histogram of the measured error rates for the different samples in the cohort after filtering.
  • the median error rate is 0.04%; 94% of samples had a measured error rate ⁇ 0.15%.
  • the error rate measurement allowed us to identify these samples and exclude these from the analysis (see below).
  • the vast majority of sequences are recipient-derived (> 90%) and therefore the net effect of assignment errors (recipient sequences assigned to donor or donor sequences assigned to the recipient) is an overestimate of the donor fraction.
  • Time courses and rejection cases Using this workflow, we first established the variability and time-dependence of the observed cfdDNA levels in the absence of biopsy- defined acute rejection.
  • Fig. 4D elevated donor DNA fractions were observed in a patient with consecutive episodes of acute cellular rejection (months 4 and 12, donor fraction 2.15% and 9.2% respectively) and antibody-mediated rejection (month 4, donor fraction 5.2 %).
  • This example illustrates the application of this technique for detection of both acute cellular and antibody-mediated rejection events.
  • cfdDNA levels were significantly higher for heart transplant recipients during acute rejection and correlated with the severity of the rejection episode as determined by biopsy (p-values 0.008 and 3.10-7, comparing biopsy grades 0 and 1 R/1A and biopsy grades 0 and > 2R/3A respectively, Mann-Whitney U test).
  • An ROC analysis of the performance of cfdDNA in distinguishing moderate-to- severe and mild rejection events yields an AUC of 0.752.
  • Organ transplants are also genome transplants, a fact that opens up the possibility of monitoring allograft injury through measurements of donor-derived cell-free DNA circulating in the recipient's plasma. Based on the present data from a single-center prospective study, we conclude that GTD is informative for monitoring acute rejection after heart transplantation and that GTD has the potential to complement or replace existing biopsy-based surveillance approaches.
  • the GTD measurement is non-invasive and is potentially compatible with high-frequency post-transplant monitoring.
  • the GTD is quantitative and therefore allows for better modulation of anti-rejection treatments.
  • the data furthermore indicate the potential for early diagnosis of acute rejection.
  • Post-transplant therapeutic protocol The post-transplant treatment protocol was previously described in detail in detail in Fan et al. (2008).
  • Biopsy surveillance All heart transplant recipients were monitored for acute rejection by surveillance endomyocardial biopsies performed at scheduled intervals after transplant: weekly during the first month, biweekly until the 3rd month, monthly until the 6th month, and then at months 9, 12, 16, 20, and 24. Biopsies were graded according to the ISHLT 2004 revised grading scale (0, 1 R, 2R, 3R).
  • Plasma processing and DNA extraction Plasma was extracted from whole blood samples as previously described ⁇ 18), and stored at -80°C. When required for analysis, plasma samples were thawed and circulating DNA was immediately extracted from 0.5-1 ml plasma using the QIAamp Circulating Nucleic Acid Kit (Qiagen).
  • Sequencing library preparation and sequencing Sequencing libraries were prepared from purified plasma DNA using the NEBNext DNA Library Prep Master Mix Set for lllumina with standard lllumina indexed adapters (IDT), or using a microfluidics-based automated library preparation platform (Mondrian ST, Ovation SP Ultralow library system).
  • Genotyping Whole blood samples were collected from the donor and recipient. DNA was purified from whole blood (DNAeasy blood and tissue kit, Qiagen) and amplified (Repli- g Mini kit, Qiagen). Genotyping was performed on lllumina Whole-genome arrays (HumanOmni2.5-8 or HumanOmnil ).
  • Custom scripts Custom scripts (written in shell, Perl or R) that were used in addition to the tools described above are made available.
  • SNP and base call filtering To reduce the number of erroneous assignments, only SNPs with high GenCall (> 0.6) and Cluster Separation (> 0.6) scores were retained. The choice of the filtering parameters was found to have only a minor influence on the detection quality.
  • the measured error rate was shown as function of the base call error probability reported by the sequencer. The measured error rate matches the predicted error rate for bases with high error rate well. At low error rate ( ⁇ 5.10 "4 ), the measured error rate was independent of the reported base call error rate, indicating that the error rate is dominated by either PCR or genotyping errors. Based on this observation we excluded base calls with reported base call error rates higher than 2.10 "3 (lllumina Q-score ⁇ 60).
  • Positron emission tomography scanning demonstrated intense fluorodeoxyglucose uptake throughout the right and left ventricular myocardium consistent with inflammatory changes that can be seen in acute rejection versus recurrent myocarditis. In view of the complicated and unusual post-transplant course experienced by this patient, we have decided not to include data for this subject in the analysis.
  • Donor-derived circulating cell-free DNA can also be used to predict a rejection event in subjects that include one or two lung transplants, which is shown in the data of FIGS. 6-9.
  • FIG. 6 depicts the fraction of donor-derived DNA in a biological sample obtained from a lung transplant recipient in the absence of rejection over a course of time.
  • FIG. 7 compares the fraction of donor-derived DNA in a biological sample obtained from a single lung transplant recipient in the absence of rejection over a course of time and a double lung transplant recipient in the absence of rejection over a course of time.
  • FIG. 8 illustrates a change in donor-derived DNA in a biological sample of a lung transplant recipient that is indicative of a rejection.
  • FIG. 9 compares the fraction of donor-derived DNA in a biological sample of lung transplant recipients treated for rejection and patients not treated for rejection.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Engineering & Computer Science (AREA)
  • Pathology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention porte sur un procédé de diagnostic non effractif pour surveiller le succès d'un implant après son implantation avec des valeurs prédictives comparables de biopsies. Les procédés de l'invention comprennent la quantification d'acide nucléique, tel que de l'ADN, acellulaire, circulant et issu du donneur (cfdDNA), afin de déterminer le succès d'un implant. La quantification peut être effectuée grâce au séquençage à haut débit du cfdDNA. Dans certains modes de réalisation, l'invention porte sur des procédés de diagnostic ou de prédiction d'état ou de résultat de greffe, comprenant les étapes consistant : (i) à utiliser un échantillon provenant d'un sujet qui a reçu un greffon d'un donneur ; (ii) à déterminer la présence ou l'absence d'un ou de plusieurs acides nucléiques provenant du greffon du donneur, ledit ou lesdits acides nucléiques provenant du donneur étant identifiés sur la base d'un profil de marqueurs prédéfini ; (iii) à effectuer une analyse bio-informatique pour améliorer la qualité de l'analyse ; (iv) à diagnostiquer ou prédire l'état ou le résultat de greffe sur la base de la présence ou de l'absence dudit ou desdits acides nucléiques.
PCT/US2014/064402 2013-11-06 2014-11-06 Adn acellulaire circulant pour le diagnostic du rejet de greffe WO2015069933A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361900912P 2013-11-06 2013-11-06
US61/900,912 2013-11-06

Publications (1)

Publication Number Publication Date
WO2015069933A1 true WO2015069933A1 (fr) 2015-05-14

Family

ID=53042091

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/064402 WO2015069933A1 (fr) 2013-11-06 2014-11-06 Adn acellulaire circulant pour le diagnostic du rejet de greffe

Country Status (1)

Country Link
WO (1) WO2015069933A1 (fr)

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017045654A1 (fr) * 2015-09-18 2017-03-23 广州华大基因医学检验所有限公司 Procédé de détermination de la proportion d'adn acellulaire source donneur dans un échantillon d'adn acellulaire récepteur
WO2018237078A1 (fr) * 2017-06-20 2018-12-27 The Medical College Of Wisconsin, Inc. Évaluation de conditions chez des sujets greffés à l'aide d'adn acellulaire spécifique d'un donneur
EP3517629A1 (fr) * 2018-01-30 2019-07-31 Myway Genetics S.r.L. Utilisation de fragments cfdna comme biomarqueurs chez des patients après une transplantation d'organes
US10385396B2 (en) 2012-04-19 2019-08-20 The Medical College Of Wisconsin, Inc. Highly sensitive surveillance using detection of cell free DNA
CN111183232A (zh) * 2017-08-17 2020-05-19 泰诊断有限公司 确定不具有供体基因型的供体无细胞dna的方法
JP2020526218A (ja) * 2017-07-14 2020-08-31 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 移植拒絶リスクを予測する新規の方法
WO2020206292A3 (fr) * 2019-04-03 2020-11-12 The Medical College Of Wisconsin, Inc. Évaluation de conditions chez des sujets greffés à l'aide d'adn acellulaire spécifique d'un donneur
JP2022500015A (ja) * 2018-09-07 2022-01-04 セクエノム, インコーポレイテッド 移植片拒絶を検出する方法およびシステム
WO2021252733A3 (fr) * 2020-06-10 2022-02-17 Bonadea Diagnostics, Llc Réaction de conversion de séquence
WO2023183826A1 (fr) * 2022-03-21 2023-09-28 Caredx, Inc. Procédés de surveillance non invasive de la santé d'un organe dans une transplantation interspécifique
US11773434B2 (en) 2017-06-20 2023-10-03 The Medical College Of Wisconsin, Inc. Assessing transplant complication risk with total cell-free DNA
US11931674B2 (en) 2019-04-04 2024-03-19 Natera, Inc. Materials and methods for processing blood samples
US11939634B2 (en) 2010-05-18 2024-03-26 Natera, Inc. Methods for simultaneous amplification of target loci
US11946101B2 (en) 2015-05-11 2024-04-02 Natera, Inc. Methods and compositions for determining ploidy
WO2024076484A1 (fr) * 2022-10-06 2024-04-11 Natera, Inc. Procédés servant à diagnostiquer et à surveiller le rejet d'une xénogreffe par mesure des protéines ou des acides nucléiques dérivés de la xénogreffe
US12020778B2 (en) 2019-03-22 2024-06-25 Natera, Inc. Methods for non-invasive prenatal ploidy calling

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120108460A1 (en) * 2010-10-26 2012-05-03 Stanford University Non-invasive fetal genetic screening by sequencing analysis
US20120196754A1 (en) * 2010-12-07 2012-08-02 Stanford University Non-invasive determination of fetal inheritance of parental haplotypes at the genome-wide scale
US20120295810A1 (en) * 2009-11-06 2012-11-22 Quake Stephen R Non-Invasive Diagnosis of Graft Rejection in Organ Transplant Patients
US20130252835A1 (en) * 2012-01-27 2013-09-26 Lian Chye Winston Koh Methods for profiling and quantitating cell-free rna

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120295810A1 (en) * 2009-11-06 2012-11-22 Quake Stephen R Non-Invasive Diagnosis of Graft Rejection in Organ Transplant Patients
US20120108460A1 (en) * 2010-10-26 2012-05-03 Stanford University Non-invasive fetal genetic screening by sequencing analysis
US20120196754A1 (en) * 2010-12-07 2012-08-02 Stanford University Non-invasive determination of fetal inheritance of parental haplotypes at the genome-wide scale
US20130252835A1 (en) * 2012-01-27 2013-09-26 Lian Chye Winston Koh Methods for profiling and quantitating cell-free rna

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DE VLAMINCK ET AL.: "Circulating cell -free DNA enables noninvasive diagnosis of heart transplant rejection", SCI TRANSL.MED., vol. 6, 18 June 2014 (2014-06-18), pages 1 - 9 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11939634B2 (en) 2010-05-18 2024-03-26 Natera, Inc. Methods for simultaneous amplification of target loci
US10472680B2 (en) 2012-04-19 2019-11-12 Medical College Of Wisconsin, Inc. Highly sensitive transplant rejection surveillance using targeted detection of donor specific cell free DNA
US10385396B2 (en) 2012-04-19 2019-08-20 The Medical College Of Wisconsin, Inc. Highly sensitive surveillance using detection of cell free DNA
US11946101B2 (en) 2015-05-11 2024-04-02 Natera, Inc. Methods and compositions for determining ploidy
WO2017045654A1 (fr) * 2015-09-18 2017-03-23 广州华大基因医学检验所有限公司 Procédé de détermination de la proportion d'adn acellulaire source donneur dans un échantillon d'adn acellulaire récepteur
CN111094593A (zh) * 2017-06-20 2020-05-01 威斯康星州立大学医学院 使用供体特异性无细胞dna来评估移植对象中的病症
JP2020529193A (ja) * 2017-06-20 2020-10-08 ザ メディカル カレッジ オブ ウィスコンシン,インコーポレイテッドThe Medical College of Wisconsin, Inc. ドナー特異的セルフリーdnaを用いる移植対象における状態の評価
WO2018237078A1 (fr) * 2017-06-20 2018-12-27 The Medical College Of Wisconsin, Inc. Évaluation de conditions chez des sujets greffés à l'aide d'adn acellulaire spécifique d'un donneur
US11773434B2 (en) 2017-06-20 2023-10-03 The Medical College Of Wisconsin, Inc. Assessing transplant complication risk with total cell-free DNA
JP2022141905A (ja) * 2017-07-14 2022-09-29 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 移植拒絶リスクを予測する新規の方法
JP2020526218A (ja) * 2017-07-14 2020-08-31 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア 移植拒絶リスクを予測する新規の方法
US11713487B2 (en) 2017-07-14 2023-08-01 The Regents Of The University Of California Methods of predicting transplant rejection risk
CN111183232A (zh) * 2017-08-17 2020-05-19 泰诊断有限公司 确定不具有供体基因型的供体无细胞dna的方法
EP3517629A1 (fr) * 2018-01-30 2019-07-31 Myway Genetics S.r.L. Utilisation de fragments cfdna comme biomarqueurs chez des patients après une transplantation d'organes
WO2019149673A1 (fr) * 2018-01-30 2019-08-08 Myway Genetics S.r.L. Utilisation de fragments d'adncf en tant que biomarqueurs chez des patients après une greffe d'organe
US11987844B2 (en) 2018-01-30 2024-05-21 4Bases Sa Use of CFDNA fragments as biomarkers in patients after organ transplantation
JP2022500015A (ja) * 2018-09-07 2022-01-04 セクエノム, インコーポレイテッド 移植片拒絶を検出する方法およびシステム
JP7485653B2 (ja) 2018-09-07 2024-05-16 セクエノム, インコーポレイテッド 移植片拒絶を検出する方法およびシステム
US12020778B2 (en) 2019-03-22 2024-06-25 Natera, Inc. Methods for non-invasive prenatal ploidy calling
WO2020206292A3 (fr) * 2019-04-03 2020-11-12 The Medical College Of Wisconsin, Inc. Évaluation de conditions chez des sujets greffés à l'aide d'adn acellulaire spécifique d'un donneur
US11931674B2 (en) 2019-04-04 2024-03-19 Natera, Inc. Materials and methods for processing blood samples
WO2021252733A3 (fr) * 2020-06-10 2022-02-17 Bonadea Diagnostics, Llc Réaction de conversion de séquence
WO2023183826A1 (fr) * 2022-03-21 2023-09-28 Caredx, Inc. Procédés de surveillance non invasive de la santé d'un organe dans une transplantation interspécifique
WO2024076484A1 (fr) * 2022-10-06 2024-04-11 Natera, Inc. Procédés servant à diagnostiquer et à surveiller le rejet d'une xénogreffe par mesure des protéines ou des acides nucléiques dérivés de la xénogreffe

Similar Documents

Publication Publication Date Title
US11390918B2 (en) Non-invasive diagnosis of graft rejection in organ transplant patients
WO2015069933A1 (fr) Adn acellulaire circulant pour le diagnostic du rejet de greffe
US11987844B2 (en) Use of CFDNA fragments as biomarkers in patients after organ transplantation
WO2015179771A2 (fr) Signatures moléculaires pour distinguer des rejets de greffe hépatique ou des lésions hépatiques

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14859765

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 17.08.2016)

122 Ep: pct application non-entry in european phase

Ref document number: 14859765

Country of ref document: EP

Kind code of ref document: A1