WO2014182598A1 - Diagnostic assay combining cellular and cell free nucleic acid - Google Patents

Diagnostic assay combining cellular and cell free nucleic acid Download PDF

Info

Publication number
WO2014182598A1
WO2014182598A1 PCT/US2014/036751 US2014036751W WO2014182598A1 WO 2014182598 A1 WO2014182598 A1 WO 2014182598A1 US 2014036751 W US2014036751 W US 2014036751W WO 2014182598 A1 WO2014182598 A1 WO 2014182598A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
nucleic acid
dna
nucleic acids
sample
Prior art date
Application number
PCT/US2014/036751
Other languages
French (fr)
Inventor
Anthony P. Shuber
Original Assignee
Physicians Choice Laboratory Services, Llc
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 Physicians Choice Laboratory Services, Llc filed Critical Physicians Choice Laboratory Services, Llc
Publication of WO2014182598A1 publication Critical patent/WO2014182598A1/en

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
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer

Definitions

  • the invention generally relates to methods for screening a body fluid for markers of disease.
  • Advanced nucleic acid screening techniques have resulted in a revolution in disease screening. It is now possible to screen a patient for a wide variety of markers indicative of disease, such as cancer, birth defects, autoimmune disease, infection, etc.
  • a sample is taken from the subject and prepared to recover nucleic acids of interest, typically DNA.
  • the samples may be cells, such as cells from a biopsy, or the samples may be fluids, such as serum from the blood.
  • a mixed sample such as whole blood, is sampled for an assay, either the cellular component (e.g., leukocytes, circulating cells) are precipitated, lysed, and sampled, or the fluid component (e.g., serum) is sampled.
  • the cellular component e.g., leukocytes, circulating cells
  • the fluid component e.g., serum
  • the standard methods for isolating nucleic acids from a mixed sample present two complications. The first is that extra preparative steps must be taken to isolate the cellular and non-cellular components. These extra steps take time, and increase the risk that the sample may be contaminated or damaged. The second is that a valuable opportunity to learn more about the sample is lost when the other portion of the mixed sample is discarded.
  • the present invention provides methods for detecting disease, e.g. cancer, using a combination of nucleic acid biomarkers in both the cellular and the cell-free portion of a body fluid sample.
  • the described methods can be used to analyze the nucleic acids in whole urine to determine whether a subject has cancer, e.g., prostate or bladder cancer.
  • the two portions of urine are separated and analyzed individually to give a unique insight into the condition of the subject.
  • the condition of the subject could not have been determined by analyzing the cellular or the cell-free component, alone.
  • all of the nucleic acids in the body fluid sample are analyzed without separating the body sample into component parts. Accordingly, the need for separation is reduced while the predictive value of the assay is advantageously increased.
  • the techniques make it easier to monitor the progression of a disease without disruptive procedures, such as biopsy.
  • the method includes collecting a urine sample and analyzing it to detect DNA biomarkers associated with bladder cancer or prostate cancer.
  • the technique can also be used to stratify patients based on their likelihood of disease.
  • the biomarkers detected include FGFR3, MMP2, TWIST1 , Vimentin and NID2.
  • epigenetic changes such as methylation of nucleic acids, can be evaluated in order to determine the condition of a subject.
  • biomarkers can be detected with single-molecule sequencing. The results of these tests are then compared to established reference ranges and are evaluated in combination to determine the likelihood of cancer.
  • biomarkers contemplated by the invention is able to provide both high positive and negative predictive values across all stages and grades of cancer, e.g., bladder cancer or prostate cancer.
  • two marker cutoffs are established; one cutoff to maximize sensitivity and negative predictive value and a second cutoff to maximize specificity and positive predictive value.
  • Marker thresholds are then set to provide maximum NPV and sensitivity, such that patients who do not have cancer might be excluded from further intervention.
  • markers cutoffs By setting marker cutoffs to high PPV and specificity, patients could also be triaged into those that might benefit from maximum intervention. Patients assessed in between those cutoffs might continue to receive standard intervention.
  • patients with all biomarkers below a predetermined cutoff are determined as having a low likelihood of cancer.
  • the screening assay includes identifying a nucleic acid sample associated with a cellular component of a body fluid as well as a nucleic acid that is not associated with a cellular component, e.g, "cell-free” or "circulating” nucleic acids.
  • the measurements are compared to threshold parameters indicative of the absence of cancer.
  • biomarkers chosen are immaterial to the operation of the invention as long as the marker is associated with the disease for which screening is being conducted.
  • exemplary biomarkers include nucleic acid biomarkers for cancer, infectious diseases, autoimmune diseases, and birth defects.
  • Biomarkers used in methods of the invention are chosen based upon their predictive value or suspected predictive value for the condition or conditions being diagnosed. Particular markers are selected based upon various diagnostic criteria, such as suspected association with disease. The number of markers chosen will depend on the number of assays performed and is at the discretion of the user. Biomarkers should be chosen that cumulatively increase the specificity/sensitivity of the assay. A panel of markers can be chosen to increase the effectiveness of diagnosis, prognosis, treatment response, and/or recurrence.
  • markers can also be chosen in consideration of the patient's history and lifestyle. For example, other diseases that the patient has, might have, or has had can effect the choice of the panel of biomarkers to be analyzed. Drugs that the patient has in his/her system may also affect biomarker selection.
  • Threshold values for any particular biomarker and associated disease are determined by reference to literature or standard of care criteria or may be determined empirically.
  • thresholds for use in association with biomarkers of the invention are based upon positive and negative predictive values associated with threshold levels of the marker. There are numerous methods for determining thresholds for use in the invention, including reference to standard values in the literature or associated standards of care. The precise thresholds chosen are immaterial as long as they have the desired association with diagnostic output.
  • the invention is applicable to diagnosis and monitoring of any disease, either in symptomatic or asymptomatic patient populations.
  • the invention can be used for diagnosis of infectious diseases, inherited diseases, and other conditions, such as disease or damage caused by drug or alcohol abuse.
  • the invention can also be applied to assess therapeutic efficacy, potential for disease recurrence or spread (e.g. metastasis).
  • Methods of the invention can be used on patients known to have a disease, or can be used to screen healthy subjects on a periodic basis. Screening can be done on a regular basis (e.g., weekly, monthly, annually, or other time interval); or as a one-time event. The outcome of the analysis may be used to alter the frequency and/or type of screening, diagnostic and/or treatment protocols. Different conditions can be screened for at different time intervals and as a function of different risk factors (e.g., age, weight, gender, history of smoking, family history, genetic risks, exposure to toxins and/or carcinogens etc., or a combination thereof). The particular screening regimen and choice of markers used in connection with the invention are determined at the discretion of the physician or technician.
  • risk factors e.g., age, weight, gender, history of smoking, family history, genetic risks, exposure to toxins and/or carcinogens etc., or a combination thereof.
  • Biomarkers associated with diseases are shown for example in Shuber (U.S. patent application number 2009/0075266), the content of which is incorporated by reference herein in its entirety.
  • the invention is especially useful in screening for cancer.
  • biomarkers associated with cancer include FGFR3, matrix metalloproteinase (MMP), neutrophil gelatinase- associated lipocalin (NGAL), MMP/NGAL complex, thymosin ⁇ 15, thymosin ⁇ 16, collagen like gene (CLG) product, prohibitin, glutathione-S-transferase, beta-5-tubulin, ubiquitin,
  • MMPs include, but are not limited to, MMP-2, MMP-9, MMP9/ GAL complex, MMP/TIMP complex, MMP/TIMP1 complex, ADAMTS-7 or ADAM- 12, among others.
  • biomarkers that have been associated with breast cancer include: ErbB2 (Her2); ESR1 ; BRCA1 ; BRCA2; p53; mdm2; cyclinl ; p27; B_Catenin; BAG 1 ; B1N1 ; BUB 1 ; C20_orfl ; CCNB 1 ; CCNE2;
  • CDC20 CDH1 ; CEGP1 ; CIAP1 ; cMYC; CTSL2; DKFZp586M07; DR5; EpCAM; EstRl ; FOXM1 ; GRB7; GSTM1 ; GSTM3; HER2; HNRPAB; ID 1 ; IGF 1 R; ITGA7; Ki_67; KNSL2; LMNB 1; MCM2; MELK; MMP 12; MMP9; MYBL2; NEK2; NME1 ; NPD009; PCNA; PR; PREP; PTTG1 ; RPLPO; Src; STK15; STMY3; SURV; TFRC; TOP2A; and TS.
  • Biomarkers associated with development of cervical cancer are shown in Patel (US 7,300,765), Pardee et al. (US 7, 153,700), Kim (US 6,905,844), Roberts et al. (US 6,316,208), Schlegel (US 2008/01 13340), Kwok et al. (US 2008/0044828), Fisher et al. (US 2005/0260566), Sastry et al. (US 2005/0048467), Lai (US 2008/03 1 1570) and Van Der Zee et al. (US 7,300,765), Pardee et al. (US 7, 153,700), Kim (US 6,905,844), Roberts et al. (US 6,316,208), Schlegel (US 2008/01 13340), Kwok et al. (US 2008/0044828), Fisher et al. (US 2005/0260566), Sastry et al. (US 2005/0048467), Lai (US 2008/03
  • biomarkers that have been associated with cervical cancer include: SC6; SIX 1 ; human cervical cancer 2 protooncogene (HCCR-2); p27; virus oncogene E6; virus oncogene E7; ⁇ 16 ⁇ 4 ⁇ ; Mem proteins (such as Mcm5); Cdc proteins; topoisomerase 2 alpha; PCNA; Ki-67; Cyclin E; p-53; PAll ; DAP-kinase; ESR1 ; APC; TIMP-3; RAR- ⁇ ; CALCA; TSLC1 ; TIMP-2; DcRl ; CUDR; DcR2; BRCA1 ; pl5; MSH2; RassflA; MLH1 ; MGMT; SOX1 ; PAX1 ; LMX1A; NKX6-1 ; WT1 ; ONECUT1 ;
  • Biomarkers associated with development of vaginal cancer are shown in Giordano (US 5,840,506), Kruk (US 2008/0009005), Hellman et al. (Br J Cancer. 100(8): 1303-1314, 2009). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with vaginal cancer include: pRb2/pl 30 and Bcl-2.
  • Biomarkers associated with development of brain cancers are shown in D'Andrea (US 2009/0081237), Murphy et al. (US 2006/0269558), Gibson et al. (US 2006/0281089), and Zetter et al. (US 2006/0160762).
  • D'Andrea US 2009/0081237)
  • Murphy et al. US 2006/0269558
  • Gibson et al. US 2006/0281089
  • Zetter et al. US 2006/0160762
  • Exemplary biomarkers that have been associated with brain cancers include: epidermal growth factor receptor (EGFR);
  • biomarkers that have been associated with renal cancers include: SC6; 36P6D5; IMP3; serum amyloid alpha; YKL-40; SC6; and carbonic anhydrase IX (CA IX).
  • Biomarkers associated with development of hepatic cancers are shown in Home et al. (US 6,974,667), Yuan et al. (US 6,897,018), Hanausek- Walaszek et al. (US 5,310,653), and Liew et al. (US 2005/0152908).
  • the contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety.
  • biomarkers that have been associated with hepatic cancers include: Tetraspan NET-6 protein; collagen, type V, alpha; glypican 3; pituitary tumor-transforming gene 1 (PTTGl); Galectin 3; solute carrier family 2, member 3, or glucose transporter 3 (GLUT3); metallothionein 1 L; CYP2A6; claudin 4; serine protease inhibitor, Kazal type I (SPINK.1); DLC-1 ; AFP; HSP70; CAP2; glypican 3; glutamine synthetase; AFP; AST and CEA.
  • Tetraspan NET-6 protein collagen, type V, alpha
  • glypican 3 pituitary tumor-transforming gene 1 (PTTGl); Galectin 3; solute carrier family 2, member 3, or glucose transporter 3 (GLUT3)
  • metallothionein 1 L metallothionein 1 L
  • CYP2A6 claudin 4
  • Biomarkers associated with development of gastric, gastrointestinal, and/or esophageal cancers are shown in Chang et al. (US 7,507,532), Bae et al. (US 7,368,255), Muramatsu et al. (US 7,090,983), Sahin et al. (US 7,527,933), Chow et al. (US 2008/0138806), Waldman et al. (US 2005/0100895), Goldenring (US 2008/0057514), An et al. (US 2007/0259368), Guilford et al. (US 2007/0184439), Wirtz et al. (US 2004/0018525), Filella et al.
  • biomarkers that have been associated with gastric, gastrointestinal, and/or esophageal cancers include: MH15 (HnlL); RUNX3; midkine;
  • Chromogranin A (CHGA); Thy-1 cell surface antigen (THY1); IPO-38; CEA; CA 19.9; GroES; TAG-72; TGM3; HE4; LGALS3; ILI RN; TRIP 13; FIGNLl ; CRIPI ; S100A4; EXOSC8; EXPI; CRCA-1 ; BRRN1 ; NELF; EREG; TMEM40; TMEM109; and guanylin cyclase C.
  • CHGA Chromogranin A
  • TTY1 Thy-1 cell surface antigen
  • CEA CA 19.9; GroES
  • TAG-72 TGM3
  • HE4 LGALS3
  • ILI RN TRIP 13
  • Biomarkers associated with development of ovarian cancer are shown in Podust et al. (US 7,510,842), Wang (US 7,348,142), O'Brien et al. (US 7,291 ,462, 6,942,978, 6,316,213, 6,294,344, and 6,268,165), Ganetta (US 7,078, 180), Malinowski et al. (US 2009/0087849), Beyer et al. (US 2009/0081685), Fischer et al. (US 2009/0075307), Mansfield et al. (US 7,510,842), Wang (US 7,348,142), O'Brien et al. (US 7,291 ,462, 6,942,978, 6,316,213, 6,294,344, and 6,268,165), Ganetta (US 7,078, 180), Malinowski et al. (US 2009/0087849), Beyer et al. (US 2009/0081685), Fischer et al. (US 2009/0075307),
  • biomarkers that have been associated with ovarian cancer include: hepcidin; tumor antigen-derived gene (TADG-15); TADG- 12; TADG-14; ZEB; PUMP- 1 ; stratum corneum chymotrytic enzyme (SCCE); NES-1 ; ⁇ ; PAI-2; cathepsin B; cathepsin L; ERCC5; MMP-2; pRb2/pl 30 gene; matrix
  • MMP-7 metalloproteinase-7
  • POP progesterone-associated endometrial protein
  • CA125 cancer antigen 125
  • CTAP3 human epididymis 4
  • PPAUR plasminogen activator urokinase receptor
  • MUC-1 FGF-2
  • cSHMT Tbx3
  • utrophin SLPI
  • osteopontin SSP1
  • MSLN mesothelin
  • SPON 1 interleukin-7
  • folate receptor 1 folate receptor 1
  • claudin 3 Biomarkers associated with development of head-and-neck and thyroid cancers are shown in Sidransky et al. (US 7,378,233), Skolnick et al. (US 5,989,815), Budiman et al. (US 2009/0075265), Hasina et al. (Cancer Research, 63 :555-559, 2003), ebebew et al. (US
  • biomarkers that have been associated with head-and-neck and thyroid cancers include: BRAF; Multiple Tumor Suppressor (MTS); PAI-2; stratifin; YWHAZ; S100- A2; S 100-A7 (psoriasin); S100-A1 1 (calgizarrin); prothymosin alpha (PTHA); L-lactate dehydrogenase A chain; glutathione S-transferase Pi; APC-binding protein EB1 ; fascin;
  • peroxiredoxin2 carbonic anhydrase I; flavin reductase; histone H3; ECM 1 ; TMPRSS4;
  • ANGPT2 T1MP1; LOXL4; p53; IL-6; EGFR; Ku70; GST-pi; and polybromo- l D.
  • biomarkers that have been associated with colorectal cancers include: 36P6D5; TTK; CDX2; NRG4; TUCAN; hMLHl ; hMSH2; M2-PK; CGA7; CJA8; PTP.alpha.; APC; p53; Ki-ras; complement C3a des- arg; alphal -antitrypsin; transferrin; MMP-1 1 ; CA-19-9; TPA; TPS; TIMP-1 ; C lOorfi;
  • CEA carcinoembryonic antigen
  • CYFRA 21- 1 a soluble fragment of cytokeratin 19
  • TAC1 carbohydrate antigen 724 (CA72-4); nicotinamide N-methyltransferase (NNMT); pyrroline-5- carboxylate reductase (PROC); S-adenosylhomocysteine hydrolase (SAHH); 1BABP-L polypeptide; and Septin 9.
  • Biomarkers associated with development of prostate cancer are shown in Sidransky (US 7,524,633), Platica (US 7,510,707), Salceda et al. (US 7,432,064 and US 7,364,862), Siegler et al. (US 7,361 ,474), Wang (US 7,348, 142), Ali et al. (US 7,326,529), Price et al. (US 7,229,770), O'Brien et al. (US 7,291 ,462), Golub et al. (US 6,949,342), Ogden et al. (US 6,841,350), An et al. (US 6, 171 ,796), Bergan et al.
  • biomarkers that have been associated with prostate cancer include: PSA; GSTP1 ; PAR; CSG; M1F; TADG-15; p53; Y L-40; ZEB; HOXC6; Pax 2; prostate-specific transglutaminase; cytokeratin 15; MEK4; MIPl- ⁇ ; fractalkine; IL- 15; ERG8; EZH2; EPC1 ; EPC2; NLGN-4Y; kallikrein 1 1 ; ABP280 (FLNA); AMACR; AR; BM28; BUB3; CaMKK; CASPASE3; CD 7; DYNAMIN; E2F1 ; E-CADHERI ; EXPORT1N; EZH2; FAS; GAS7; GS28; ICBP90; ITGA5; JAGGED 1 ; JAM1 ; ANADAPTIN; LF6; KRIP1 ; LAP2; MCAM; IB 1 (MKI67); MTA
  • Biomarkers associated with development of pancreatic cancer are shown in Sahin et al. (US 7,527,933), Rataino et al. (US 7,507,541), Schleyer et al. (US 7,476,506), Domon et al. (US 7,473,531), McCaffey et al. (US 7,358,231), Price et al. (US 7,229,770), Chan et al. (US 2005/009561 1), Mitchl et al. (US 2006/0258841), and Faca et al. (PLoS Med 5(6):el23, 2008).
  • the contents of each of the articles, patents, and patent applications are incorpo rated by reference herein in their entirety.
  • Exemplary biomarkers that have been associated with pancreatic cancer include: CA19.9; 36P6D5; NRG4; ASCT2; CCR7; 3C4-Ag; KLK1 1 ;
  • Biomarkers associated with development of lung cancer are shown in Sahin et al. (US 7,527,933), Hutteman (US 7,473,530), Bae et al. (US 7,368,255), Wang (US 7,348, 142), Marin et al. (US 7,332,590), Gure et al. (US 7,314,721), Patel (US 7,300,765), Price et al. (US 7,527,933), Hutteman (US 7,473,530), Bae et al. (US 7,368,255), Wang (US 7,348, 142), Switzerland et al. (US 7,332,590), Gure et al. (US 7,314,721), Patel (US 7,300,765), Price et al. (US 7,527,933), Hutteman (US 7,473,530), Bae et al. (US 7,368,255), Wang (US 7,348, 142), Marin (US 7,348, 142), Switzerland et
  • biomarkers that have been associated with lung cancer include: COX-2; COX4-2; RUNX3; aldoketoreductase family 1 , member B 10; peroxiredoxin 1 (PRDX1); T F receptor superfamily member 18; small proline-rich protein 3 (SPRR3); SOX1 ; SC6; TADG- 15; YKL40; midkine; DAP-kinase;
  • HOXA9 SCCE; STX1A; HIF1A; CCT3; HLA-DPB1 ; MAFK; RNF5; KIF1 1 ; GHSRl b;
  • Biomarkers associated with development of skin cancer are shown in Roberts et al. (US 6,316,208), Polsky (US 7,442,507), Price et al. (US 7,229,770), Genetta (US 7,078, 180), Carson et al. (US 6,316,208), Polsky (US 7,442,507), Price et al. (US 7,229,770), Genetta (US 7,078, 180), Carson et al. (US 6,316,208), Polsky (US 7,442,507), Price et al. (US 7,229,770), Genetta (US 7,078, 180), Carson et al. (US
  • Cystatin B Chaperonin 10; Profilin; BRAF; YKL-40; DDX48; erbB3 -binding protein; biliverdin reductase; PLAB; LICAM; SAA; CRP; SOX9; MMP2; CD 10; and ZEB.
  • Biomarkers associated with development of multiple myeloma are shown in Coignet (US 7,449,303), Shaughnessy et al. (US 7,308,364), Seshi (US 7,049,072), and Shaughnessy et al. (US 2008/0293578, US 2008/0234139, and US 2008/0234138).
  • the contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety.
  • Exemplary biomarkers that have been associated with multiple myeloma include: JAG2;
  • CCND1 CCND1 ; MAF; MAFB; MMSET; CST6; RAB7L1 ; MAP4K3; HRASLS2; TRAIL; IG; FGL2; GNG1 1 ; MCM2; FLJ10709; TRIM13; NADSYN1 ; TRIM22; AGRN; CENTD2; SESN1 ;
  • Biomarkers associated with development of leukemia are shown in Ando et al. (US 7,479,371), Coignet (US 7,479,370 and US 7,449,303), Davi et al. (US 7,416,851), Chiorazzi (US 7,316,906), Seshi (US 7,049,072), Van Baren et al. (US 6, 130,052), Taniguchi (US 5,643,729), Hans et al. (US 2009/0131353), and Van Bockstaele et al. (Blood Rev. 23(l):25-47, 2009).
  • biomarkers that have been associated with leukemia include: SCGF; JAG2; LPL; ADAM29; PDE; Cryptochrome-1 ; CD49d; ZAP-70; PRAME; WT1 ; CD15; CD33; and CD38.
  • Biomarkers associated with development of lymphoma are shown in Ando et al. (US 7,479,371), Levy et al. (US 7,332,280), and Arnold (US 5,858,655). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with lymphoma include: SCGF; L 02; BCL6; FN1 ; CCND2; SCYA3; BCL2; CD79a; CD7; CD25; CD45RO; CD45RA; and PRAD1 cyclin.
  • Biomarkers associated with development of bladder cancer are shown in Price et al. (US 7,229,770), Orntoft (US 6,936,417), Haak-Frendscho et al. (US 6,008,003), Feinstein et al. (US 6,998,232), Elting et al. (US 2008/031 1604), and Wewer et al. (2009/0029372).
  • the contents of each of the patent applications and each of these patents are incorporated by reference herein in their entirety.
  • biomarkers that have been associated with bladder cancer include: FGFR3, NT-3; NGF; GDNF; Y L-40; p53; pRB; p21 ; p27; cyclin El ; i67; Fas; urothelial carcinoma-associated 1 ; human chorionic gonadotropin beta type II; insulin-like growth factor- binding protein 7; sorting nexin 16; chondroitin sulfate proteoglycan 6; cathepsin D;
  • chromodomain helicase DNA-binding protein 2 nell-like 2; tumor necrosis factor receptor superfamily member 7; cytokeratin 18 (C 18); ADAM8; ADAM10; ADAM12; Matrix Metalloproteinase-2 (MMP-2); MMP-9; KAI1 ; and bladder tumor fibronectin (BTF).
  • nucleic acids and proteins associated with a certain cancer vary with respect to the genetic, biochemical, or molecular alterations that associate the nucleic acid or protein with cancer.
  • the cancer causing alterations can include abnormal protein expressions, sequence mutations, methylation patterns, and loss of heterozygosity. Because multiple alterations can be linked to cancer, methods of the invention realize that there is great clinical value in assaying for multiple genetic characteristics across the plurality of biomarkers.
  • the invention involves obtaining a urine or tissue sample, conducting an assay on the urine or tissue sample to look for a nucleic acid mutation, loss of heterozygosity, and an abnormal protein level, and determining whether the sample is positive or negative for cancer based on the assay. By detecting different alterations in a signal assay, the result is a multimodal analysis that has greater sensitivity and specificity with regard to the diagnosis and
  • Methods of the invention provide for conducting an assay on a plurality of biomarkers to look for characteristics such as a nucleic acid mutation, a loss of heterozygosity, an abnormal protein level, gene expression patterns, an abnormal methylation pattern, and any other characteristic indicative of cancer.
  • the presence or absence of one or more characteristic is indicative of a positive result for the cancer to be diagnosed.
  • the type of characteristic looked for in the plurality of biomarkers is based on the cancer being diagnosed.
  • characteristics associated with bladder cancer include nucleic acid mutations, loss of heterozygosity, abnormal protein levels, and hypermethylation, whereas other cancer types might only be associated with abnormal protein level and hypermethylation patterns. Below the type of characteristics in proteins and nucleic acids that are suitable for use in methods of the invention are exemplified.
  • Nucleic acid biomarkers are often associated with nucleic acid mutations, which include additions, deletions, insertions, rearrangements, inversions, transitions, transversions, frameshift mutations, nonsense mutations, missense mutations, single nucleotide polymorphisms (SNP) and substitutions of two or more nucleotides within a sequence but not to the extent of large chromosomal sequence changes.
  • SNPs are a type of genomic subtle sequence change that occurs when a single nucleotide replaces another within the sequence.
  • Alterations in chromosome numbers include additions, deletions, inversions, translocations, copy number variations, and substitutions of chromosomes within a sequence.
  • Loss of heterozygosity results from a deletion or other mutational event within a normal allele at a particular locus heterozygous for a deleterious mutant allele and the normal allele.
  • the mutation in the normal allele renders the cell either hemizygous (one deleterious allele and one deleted allele) or homozygous for the deleterious allele.
  • the loss of the normal allele is the LOH and may be a genetic determinant in the development of cancer.
  • loss of heterozygosity in the p53 gene is associated with bladder cancer. See Oka et al., "Detection of loss of heterozygosity in the p53 gene in renal cell carcinoma and bladder cancer using the polymerase chain reaction.” Molecular Carcinogenesis: Volume 4, Issue 1 , 2006.
  • the level of protein biomarkers in the sample is analyzed in the multi-analyte screening assay to determine if there is an abnormal protein level in the sample.
  • Protein biomarkers are generally considered quantitative biomarkers for which a level or amount of the biomarker present in comparison to a reference level or amount indicates a clinical status.
  • matrix metalloproteinases such as MMP-2, MMP-9, and metalloproteases, such as ADAM- 12
  • MMPs have been shown to be key regulators of tumor growth, angiogenesis and metastasis formation. Increased MMP expression is required for tumors to grown into the surrounding tissue and for dissemination of metastatic cells into the vasculature and distant sites.
  • MMPs in the urine of cancer patients has been shown to correlate with disease status in a variety of cancers, including bladder cancer.
  • Biologically active MMP-2 and MMP-9 are found at higher levels and at greater frequency in urine of cancer patients than in healthy controls.
  • ADAM 12 is expressed in higher levels in cancer subjects than in healthy controls and is described in commonly-owned U.S. Application No. 12/120,544.
  • methods of the invention optionally include screening for the presence or absence of a methylation pattern in nucleic acid biomarkers, which includes screening nucleic acids for de-methylation, methylation, hypomethylation and hypermethylation.
  • DNA methylation is an important regulator of gene transcription and a large body of evidence has demonstrated that aberrant DNA methylation is associated with unscheduled gene silencing, and the genes with high levels of 5-methylcytosine in their promoter region are transcriptionally silent.
  • Aberrant DNA methylation patterns have been associated with a large number of human malignancies and found in two distinct forms: hypermethylation and hypomethylation compared to normal tissue.
  • Hypermethylation is one of the major epigenetic modifications that repress transcription via promoter region of tumor suppressor genes.
  • Hypermethylation typically occurs at CpG islands in the promoter region and is associated with gene inactivation. Global hypomethylation has also been shown to be causally related to the development and progression of cancer through different mechanisms. For example, a hypermethylation pattern of TWIST1, NID2, and vimentin detected in urine samples is indicative of a positive result for bladder cancer. See Renard I et al., Eur Urol. 2010; 58(1):96-104.
  • the multi-analyte screening assay includes screening for gene expression of nucleic acids.
  • Nucleic acid biomarkers associated with gene expression are generally considered quantitative biomarkers for which a level or amount of the biomarker present in comparison to a reference level or amount indicates a clinical status.
  • genes that exhibited significant over-expression in bladder cancer v.s. normal tissue include VEGFA, pl6INK4A, p53, EGFR, EGF, Ki-67, RAS, NRAS, and cyclin Dl . See, e.g.
  • Nucleic acid biomarkers generally produce a binary result, i.e., presence or absence of an alteration or characteristic in the sample as compared to a healthy control is indicative of a clinical status.
  • Protein biomarkers are generally considered quantitative biomarkers for which a level or amount of the biomarker present in comparison to a reference level or amount indicates a clinical status.
  • threshold values for any particular biomarker and associated disease may be determined by reference to literature or standard of care criteria or may be determined empirically.
  • Amplification refers to production of additional copies of a nucleic acid sequence. See for example, Dieffenbach and Dveksler, PCR Primer, a Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y. (1995), the contents of which is hereby incorporated by reference in its entirety.
  • the amplification reaction may be any amplification reaction known in the art that amplifies nucleic acid molecules, such as polymerase chain reaction, nested polymerase chain reaction, polymerase chain reaction-single strand conformation polymorphism, ligase chain reaction, strand displacement amplification and restriction fragments length polymorphism.
  • the target nucleic acid and the nucleic acid ligand are PCR amplified.
  • PCR refers to methods by K. B. Mullis (U.S. patent numbers 4,683, 195 and 4,683,202, hereby incorporated by reference) for increasing concentration of a segment of a target sequence in a mixture of genomic DNA without cloning or purification.
  • the process for amplifying the target nucleic acid sequence and nucleic acid ligand includes introducing an excess of oligonucleotide primers that bind the nucleic acid and the nucleic acid ligand, followed by a precise sequence of thermal cycling in the presence of a DNA polymerase.
  • the primers are complementary to their respective strands of the target nucleic acid and nucleic acid ligand.
  • the mixture of primers are annealed to their complementary sequences within the target nucleic acid and nucleic acid ligand.
  • the primers are extended with a polymerase so as to form a new pair of complementary strands.
  • the steps of denaturation, primer annealing and polymerase extension can be repeated many times (i.e., denaturation, annealing, and extension constitute one cycle; there can be numerous cycles) to obtain a high concentration of an amplified segment of a desired target and nucleic acid ligand.
  • the length of the amplified segment of the desired target and nucleic acid ligand is determined by relative positions of the primers with respect to each other, and therefore, this length is a controllable parameter.
  • PCR With PCR, it is possible to amplify a single copy of a specific target sequence in genomic DNA to a level that can be detected by several different methodologies (e.g., staining, hybridization with a labeled probe, incorporation of biotinylated primers followed by avidin- enzyme conjugate detection, incorporation of 32P-labeled deoxynucleotide triphosphates, such as dCTP or dATP, into the amplified segment).
  • staining hybridization with a labeled probe
  • biotinylated primers followed by avidin- enzyme conjugate detection incorporation of 32P-labeled deoxynucleotide triphosphates, such as dCTP or dATP, into the amplified segment.
  • 32P-labeled deoxynucleotide triphosphates such as dCTP or dATP
  • the target nucleic acid and nucleic acid ligand can be detected using detectably labeled probes.
  • Nucleic acid probe design and methods of synthesizing oligonucleotide probes are known in the art. See, e.g., Sambrook et al., DNA microarray: A Molecular Cloning Manual, Cold Spring Harbor, N.Y., (2003) or Maniatis, et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, N.Y., (1982), the contents of each of which are herein incorporated by reference herein in their entirety. Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd Ed.), Vols.
  • Probes suitable for use in the present invention include those formed from nucleic acids, such as RNA and/or DNA, nucleic acid analogs, locked nucleic acids, modified nucleic acids, and chimeric probes of a mixed class including a nucleic acid with another organic component such as peptide nucleic acids. Probes can be single stranded or double stranded. Exemplary nucleotide analogs include phosphate esters of deoxyadenosine, deoxycytidine, deoxyguanosine, deoxythymidine, adenosine, cytidine, guanosine, and uridine.
  • non-natural nucleotides include a xanthine or hypoxanthine; 5-bromouracil, 2-aminopurine, deoxyinosine, or methylated cytosine, such as 5-methylcytosine, and N4-methoxydeoxycytosine.
  • bases of polynucleotide mimetics such as methylated nucleic acids, e.g., 2'-0-methRNA, peptide nucleic acids, modified peptide nucleic acids, and any other structural moiety that can act substantially like a nucleotide or base, for example, by exhibiting base-complementarity with one or more bases that occur in DNA or RNA.
  • probes may be of any length.
  • probes may be as few as 5 nucleotides, or as much as 5000 nucleotides.
  • Exemplary probes are 5-mers, 10-mers, 15-mers, 20-mers, 25-mers, 50-mers, 100-mers, 200- mers, 500-mers, 1000-mers, 3000-mers, or 5000-mers. Methods for determining an optimal probe length are known in the art. See, e.g., Shuber, U.S. Patent Number 5,888,778, hereby incorporated by reference in its entirety.
  • Probes used for detection may include a detectable label, such as a radiolabel, fluorescent label, or enzymatic label. See for example Lancaster et al., U.S. Patent Number 5,869,717, hereby incorporated by reference.
  • the probe is fluorescently labeled. Fluorescently labeled nucleotides may be produced by various techniques, such as those described in Kambara et al., Bio/Technol., 6:816-21 , (1988); Smith et al., Nucl. Acid Res., 13:2399-2412, (1985); and Smith et al., Nature, 321 : 674-679, (1986), the contents of each of which are herein incorporated by reference in their entirety.
  • the fluorescent dye may be linked to the deoxyribose by a linker arm that is easily cleaved by chemical or enzymatic means.
  • linker arm that is easily cleaved by chemical or enzymatic means.
  • There are numerous linkers and methods for attaching labels to nucleotides as shown in Oligonucleotides and Analogues: A Practical Approach, IRL Press, Oxford, (1991); Zuckerman et al., Polynucleotides Res., 15: 5305-5321, ( 1987); Sharma et al., Polynucleotides Res., 19:3019, (1991); Giusti et al., PCR Methods and Applications, 2:223-227, (1993); Fung et al. (U.S. Patent Number 4,757,141); Stabinsky (U.S.
  • Patent Number 4,739,044) ; Agrawal et al., Tetrahedron Letters, 31 : 1543-1546, (1990); Sproat et al., Polynucleotides Res., 15:4837, (1987); and Nelson et al., Polynucleotides Res., 17:7187-7194, (1989), the contents of each of which are herein incorporated by reference in their entirety. Extensive guidance exists in the literature for derivatizing fluorophore and quencher molecules for covalent attachment via common reactive groups that may be added to a nucleotide.
  • linking moieties and methods for attaching fluorophore moieties to nucleotides also exist, as described in Oligonucleotides and Analogues, supra; Guisti et al., supra; Agrawal et al, supra; and Sproat et al., supra
  • the detectable label attached to the probe may be directly or indirectly detectable.
  • the exact label may be selected based, at least in part, on the particular type of detection method used.
  • Exemplary detection methods include radioactive detection, optical absorbance detection, e.g., UV-visible absorbance detection, optical emission detection, e.g., fluorescence; phosphorescence or chemiluminescence; Raman scattering.
  • Preferred labels include optically-detectable labels, such as fluorescent labels.
  • fluorescent labels include, but are not limited to, 4-acetamido-4'-isothiocyanatostilbene-2,2'disulfonic acid; acridine and derivatives: acridine, acridine isothiocyanate; 5-(2'-aminoethyl)aminonaphthalene-l -sulfonic acid (EDANS); 4-amino-N-[3-vinylsulfonyl)phenyl]naphthalimide-3,5 disulfonate; N-(4-anilino- l -naphthyl)maleimide; anthranilamide; BODIPY; alexa; fluorescien; conjugated multi-dyes; Brilliant Yellow; coumarin and derivatives; coumarin, 7-amino-4-methylcoumarin (AMC, Coumarin 120), 7-amino-4-trifluoromethylcouluarin (Coumaran 151); cyanine dyes
  • diethylenetriamine pentaacetate 4,4'-diisothiocyanatodihydro-stilbene-2,2'-disulfonic acid; 4,4'- diisothiocyanatostilbene-2,2'-disulfonic acid; 5-[dimethylamino]naphthalene-l-sulfonyl chloride (DNS, dansylchloride); 4-dimethylaminophenylazophenyl-4'-isothiocyanate (DABITC); eosin and derivatives; eosin, eosin isothiocyanate, erythrosin and derivatives; erythrosin B, erythrosin, isothiocyanate; ethidium; fluorescein and derivatives; 5-carboxyfluorescein (FAM), 5-(4,6- dichlorotriazin-2-yl)aminofluorescein (DTAF), 2',7'-dimethoxy-4'5'-
  • N,N,N',N'tetramethyl-6-carboxyrhodamine TAMRA
  • tetramethyl rhodamine TAMRA
  • tetramethyl rhodamine TAMRA
  • TRITC tetramethyl rhodamine isothiocyanate
  • riboflavin rosolic acid; terbium chelate derivatives; Atto dyes, Cy3; Cy5; Cy5.5; Cy7; IRD 700; IRD 800; La Jolta Blue; phthalo cyanine; and naphthalo cyanine.
  • Labels other than fluorescent labels are contemplated by the invention, including other optically-detectable labels.
  • Detection of a bound probe may be measured using any of a variety of techniques dependent upon the label used, such as those known to one of skill in the art.
  • Exemplary detection methods include radioactive detection, optical absorbance detection, e.g., UV-visible absorbance detection, optical emission detection, e.g., fluorescence or chemiluminescence.
  • Devices capable of sensing fluorescence from a single molecule include scanning tunneling microscope (siM) and the atomic force microscope (AFM).
  • Hybridization patterns may also be scanned using a CCD camera (e.g., Model TE/CCD512SF, Princeton Instruments, Trenton, N.J.) with suitable optics (Ploem, in Fluorescent and Luminescent Probes for Biological Activity Mason, T. G. Ed., Academic Press, Landon, pp. 1 -1 1 (1993)), such as described in Yershov et al., Proc. Natl. Acad. Sci. 93:4913 (1996), or may be imaged by TV monitoring.
  • a CCD camera e.g., Model TE/CCD512SF, Princeton Instruments, Trenton, N.J.
  • suitable optics Ploem, in Fluorescent and Luminescent Probes for Biological Activity Mason, T. G. Ed., Academic Press, Landon, pp. 1 -1 1 (1993)
  • Yershov et al. Proc. Natl. Acad. Sci. 93:4913 (1996), or may be
  • a phosphorimager device For radioactive signals, a phosphorimager device can be used (Johnston et al., Electrophoresis, 13:566, 1990; Drmanac et al., Electrophoresis, 13:566, 1992; 1993).
  • Other commercial suppliers of imaging instruments include General Scanning Inc., (Watertown, Mass. on the World Wide Web at genscan.com), Genix Technologies (Waterloo, Ontario, Canada; on the World Wide Web at confocal.com), and Applied Precision Inc.
  • the target nucleic acid or nucleic acid ligand or both are quantified using methods known in the art.
  • a preferred method for quantitation is quantitative polymerase chain reaction (QPCR).
  • QPCR refers to a PCR reaction performed in such a way and under such controlled conditions that the results of the assay are quantitative, that is, the assay is capable of quantifying the amount or concentration of a nucleic acid ligand present in the test sample.
  • QPCR is a technique based on the polymerase chain reaction, and is used to amplify and simultaneously quantify a targeted nucleic acid molecule.
  • QPCR allows for both detection and quantification (as absolute number of copies or relative amount when normalized to DNA input or additional normalizing genes) of a specific sequence in a DNA sample.
  • the procedure follows the general principle of PCR, with the additional feature that the amplified DNA is quantified as it accumulates in the reaction in real time after each amplification cycle.
  • QPCR is described, for example, in Kurnit et al. (U.S. Patent Number 6,033,854), Wang et al. (U.S. Patent Numbers 5,567,583 and 5,348,853), Ma et al.
  • Two common methods of quantification are: (1) use of fluorescent dyes that intercalate with double-stranded DNA, and (2) modified DNA oligonucleotide probes that fluoresce when hybridized with a complementary DNA.
  • a DNA-binding dye binds to all double-stranded (ds)DNA in PCR, resulting in fluorescence of the dye.
  • An increase in DNA product during PCR therefore leads to an increase in fluorescence intensity and is measured at each cycle, thus allowing DNA concentrations to be quantified.
  • the reaction is prepared similarly to a standard PCR reaction, with the addition of fluorescent (ds)DNA dye.
  • the reaction is run in a thermocycler, and after each cycle, the levels of fluorescence are measured with a detector; the dye only fluoresces when bound to the (ds)DNA (i.e., the PCR product).
  • the ds double-stranded
  • (ds)DNA concentration in the PCR can be determined. Like other real-time PCR methods, the values obtained do not have absolute units associated with it. A comparison of a measured DNA/RNA sample to a standard dilution gives a fraction or ratio of the sample relative to the standard, allowing relative comparisons between different tissues or experimental conditions. To ensure accuracy in the quantification, it is important to normalize expression of a target gene to a stably expressed gene. This allows for correction of possible differences in nucleic acid quantity or quality across samples.
  • the second method uses sequence-specific RNA or DNA-based probes to quantify only the DNA containing the probe sequence; therefore, use of the reporter probe significantly increases specificity, and allows for quantification even in the presence of some non-specific DNA amplification. This allows for multiplexing, i.e., assaying for several genes in the same reaction by using specific probes with differently colored labels, provided that all genes are amplified with similar efficiency.
  • This method is commonly carried out with a DNA-based probe with a fluorescent reporter (e.g. 6-carboxyfluorescein) at one end and a quencher (e.g., 6-carboxy- tetramethylrhodamine) of fluorescence at the opposite end of the probe.
  • a fluorescent reporter e.g. 6-carboxyfluorescein
  • a quencher e.g., 6-carboxy- tetramethylrhodamine
  • An increase in the product targeted by the reporter probe at each PCR cycle results in a proportional increase in fluorescence due to breakdown of the probe and release of the reporter.
  • the reaction is prepared similarly to a standard PCR reaction, and the reporter probe is added. As the reaction commences, during the annealing stage of the PCR, both probe and primers anneal to the DNA target. Polymerization of a new DNA strand is initiated from the primers, and once the polymerase reaches the probe, its 5 '-3 '-exonuclease degrades the probe, physically separating the fluorescent reporter from the quencher, resulting in an increase in fluorescence. Fluorescence is detected and measured in a real-time PCR thermocycler, and geometric increase of fluorescence corresponding to exponential increase of the product is used to determine the threshold cycle in each reaction.
  • the QPCR reaction uses fluorescent TaqmanTM methodology and an instrument capable of measuring fluorescence in real time (e.g., ABI Prism 7700 Sequence Detector; see also PE Biosystems, Foster City, Calif.; see also Gelfand et al., (U.S. Patent Number 5,210,015), the contents of which is hereby incorporated by reference in its entirety).
  • the TaqmanTM reaction uses a hybridization probe labeled with two different fluorescent dyes. One dye is a reporter dye (6-carboxyfluorescein), the other is a quenching dye (6-carboxy- tetramethylrhodamine). When the probe is intact, fluorescent energy transfer occurs and the reporter dye fluorescent emission is absorbed by the quenching dye.
  • the fluorescent hybridization probe is cleaved by the 5 '-3' nucleoiytic activity of the DNA polymerase. On cleavage of the probe, the reporter dye emission is no longer transferred efficiently to the quenching dye, resulting in an increase of the reporter dye fluorescent emission spectra.
  • the nucleic acid ligand of the present invention is quantified by performing QPCR and determining, either directly or indirectly, the amount or concentration of nucleic acid ligand that had bound to its probe in the test sample.
  • the amount or concentration of the bound probe in the test sample is generally directly proportional to the amount or concentration of the nucleic acid ligand quantified by using QPCR. See for example Schneider et al., U.S. Patent Application Publication Number 2009/0042206, Dodge et al., U.S. Patent Number 6,927,024, Gold et al., U.S. Patent Numbers 6,569,620, 6,716,580, and 7,629, 151 , Cheronis et al., U.S. Patent Number 7,074,586, and Ahn et al., U.S. Patent Number 7,642,056, the contents of each of which are herein incorporated by reference in their entirety.
  • the presence of the nucleic acid in the analyzed sample directly correlates to the presence of the target protein in that sample.
  • the amount of nucleic acid present in the sample correlates to the signal intensity following the conduction of the PCR-based methods.
  • the signal intensity of PCR depends upon the number of PCR cycles performed and/or the starting concentration of the nucleic acid. Since the sequence of the target protein is known to generate the nucleic acid, detection of that specific nucleic acid correlates to the presence of the target protein. Similarly, detection of the amplified target nucleic acid indicates the presence of the target nucleic acid in the sample analyzed.
  • one method for detection and quantification of amplified nucleic acid or target nucleic acid results from the presence of a fluorogenic probe.
  • the probe which is specific for the nucleic acid, has a 6- carboxyfluorescein (FAM) moiety covalently bound to the 5-'end and a 6- carboxytetramethylrhodamine (TAMRA) or other fluorescent-quenching dye (easily prepared using standard automated DNA synthesis) present on the 3'-end, along with a 3'-phosphate to prevent elongation.
  • FAM 6- carboxyfluorescein
  • TAMRA 6- carboxytetramethylrhodamine
  • the probe is added with 5'-nuclease to the PCR assays, such that 5'-nuclease cleavage of the probe-nucleic acid duplex results in release of the 5'-bound FAM moiety from the oligonucleotide probe.
  • 5'-nuclease cleavage of the probe-nucleic acid duplex results in release of the 5'-bound FAM moiety from the oligonucleotide probe.
  • cycler/laser/detector/software system such as an ABI 7700 Sequence Detector (Applied
  • the ABI instrument has the advantage of allowing analysis and display of quantification in less than 60s upon termination of the amplification reactions.
  • Both detection systems employ an internal control or standard wherein a second nucleic acid sequence utilizing the same primers for amplification but having a different sequence and thus different probe, is amplified, monitored and quantitated simultaneously as that for the desired target molecule. See for example, "A Novel Method for Real Time Quantitative RT-PCR," Gibson, U. et. al., 1996, Genome Res. 6:995-1001 ; Piatak, M. et. al., 1993, BioTechniques 14:70-81 ;
  • the primers used for amplification contain molecular energy transfer (MET) moieties, specifically fluorescent resonance energy transfer (FRET) moieties, whereby the primers contain both a donor and an acceptor molecule.
  • MET molecular energy transfer
  • FRET fluorescent resonance energy transfer
  • the FRET pair typically contains a fluorophore donor moiety such as 5-carboxyfluorescein (FAM) or 6-carboxy-4,5-dichloro-2,7- dimethoxyfluorescein (JOE), with an emission maximum of 525 or 546 nm, respectively, paired with an acceptor moiety such as N ⁇ TSi'N'-tetramethyl-6-carboxyrhodamine (TAMRA), 6- carboxy-X-rhodamine (ROX) or 6-carboxyrhodamine (R6G), all of which have excitation maximum of 514 nm.
  • FAM fluorophore donor moiety
  • FEM 6-carboxy-4,5-dichloro-2,7- dimethoxyfluorescein
  • the primer may be a hairpin such that the 5'-end of the primer contains the FRET donor, and the 3'-end (based-paired to the 5'-end to form the stem region of the hairpin) contains the FRET acceptor, or quencher.
  • the two moieties in the FRET pair are separated by approximately 15-25 nucleotides in length when the hairpin primer is linearized. While the primer is in the hairpin conformation, no fluorescence is detected. Thus, fluorescence by the donor is only detected when the primer is in a linearized conformation, i.e. when it is incorporated into a double-stranded amplification product.
  • Such a method allows direct quantification of the amount of nucleic acid bound to target molecule in the sample mixture, and this quantity is then used to determine the amount of target molecule originally present in the sample. See for example, Nazarenko, I. A. et al., U.S. Pat. No. 5,866,336, the contents of which is incorporated by reference in its entirety.
  • a TaqManTM probe selects a TaqManTM probe based upon the sequence of the nucleic acid to be quantified and generally includes a 5'-end fluor, such as 6-carboxyfluorescein, for example, and a 3'-end quencher, such as, for example, a 6-carboxytetramethylfluorescein, to generate signal as the nucleic acid sequence is amplified using PCR.
  • a 5'-end fluor such as 6-carboxyfluorescein, for example
  • a 3'-end quencher such as, for example, a 6-carboxytetramethylfluorescein
  • the amount or concentration of an nucleic acid affinity complex is determined using an intercalating fluorescent dye during the replicative process.
  • the intercalating dye such as, for example, SYBRTM green, generates a large fluorescent signal in the presence of double-stranded DNA as compared to the fluorescent signal generated in the presence of single-stranded DNA.
  • SYBRTM green As the double-stranded DNA product is formed during PCR, the signal produced by the dye increases. The magnitude of the signal produced is dependent upon both the number of PCR cycles and the starting concentration of the nucleic acid.
  • Nucleic acids and proteins may be obtained by methods known in the art. Generally, nucleic acids can be extracted from a biological sample by a variety of techniques such as those described by Maniatis, et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, N.Y., pp. 280-281 , (1982), the contents of which is incorporated by reference herein in its entirety. Generally, proteins can be extracted from a biological sample by a variety of techniques such as 2-D electrophoresis, isoelectric focusing, and SDS Slab Gel Electrophoresis. See for example O'Farrell, J. Biol. Chem., 250: 4007-4021 (1975), Sambrook, J.
  • antibodies with a unique oligonucleotide tag are added to the sample to bind a target protein and detection of the oligonucleotide tag results in detection of the protein.
  • the target protein is exposed to an antibody that is coupled to an oligonucleotide tag of a known sequence.
  • the antibody specifically binds the protein, and then PCR is used to amplify the oligonucleotide coupled to the antibody.
  • the identity of the target protein is determined based upon the sequence of the oligonucleotide attached to the antibody and the presence of the oligonucleotide in the sample. In this embodiment of the invention, different antibodies specific for the target protein are used.
  • Each antibody is coupled to a unique oligonucleotide tag of known sequence. Therefore, more than one target protein can be detected in a sample by x identifying the unique oligonucleotide tag attached to the antibody. See for example Kahvejian, U.S. Patent Application Publication Number 2007/0020650, hereby incorporated by reference.
  • antibodies with a unique nucleotide tag are added to the sample to bind the target nucleic acid.
  • different antibodies specific for the target nucleic acid are used, therefore, more than one target nucleic acid can be detected in a sample by identifying the unique oligonucleotide tag attached.
  • Detection of the nucleotide tag may be done by methods known in the art, such as PCR, QPCR, fluorescent labeling, radiolabeling, biotinylation, Sanger sequencing, sequencing by synthesis, or Single Molecule Real Time Sequencing methods.
  • Single molecule sequencing methods see for example, Lapidus, U.S. Patent Number 7,666,593, Quake et al., U.S. Patent Number 7,501,245, and Lapidus et al., U.S. Patent Numbers 7,169,560 and 7,491 ,498, the contents of each of which are herein incorporated by reference.
  • Antibodies for use in the present invention can be generated by methods well known in the art. See, for example, E. Harlow and D. Lane, Antibodies, a Laboratory Model, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (1988), the contents of which are hereby incorporated by reference in their entirety. In addition, a wide variety of antibodies are available commercially.
  • the antibody can be obtained from a variety of sources, such as those known to one of skill in the art, including but not limited to polyclonal antibody, monoclonal antibody, monospecific antibody, recombinantly expressed antibody, humanized antibody, plantibodies, and the like; and can be obtained from a variety of animal species, including rabbit, mouse, goat, rat, human, horse, bovine, guinea pig, chicken, sheep, donkey, human, and the like.
  • a wide variety of antibodies are commercially available and a custom-made antibody can be obtained from a number of contract labs.
  • the target nucleic acid or nucleic acid ligand or both are detected using sequencing.
  • the nucleic acid/protein complex may be dissociated, releasing the nucleic acid for the sequencing reaction. Sequencing-by-synthesis is a common technique used in next generation procedures and works well with the instant invention.
  • sequencing involves hybridizing a primer to a template to form a template/primer duplex, contacting the duplex with a polymerase in the presence of a detectably-labeled nucleotides under conditions that permit the polymerase to add nucleotides to the primer in a template-dependent manner. Signal from the detectable label is then used to identify the incorporated base and the steps are sequentially repeated in order to determine the linear order of nucleotides in the template.
  • exemplary detectable labels include radiolabels, florescent labels, enzymatic labels, etc.
  • the detectable label may be an optically detectable label, such as a fluorescent label.
  • exemplary fluorescent labels include cyanine, rhodamine, fluorescien, coumarin, BODIPY, alexa, or conjugated multi-dyes. Numerous techniques are known for detecting sequences and some are exemplified below. However, the exact means for detecting and compiling sequence data does not affect the function of the invention described herein.
  • the target nucleic acids, nucleic acid ligands, or both are detected using single molecule sequencing.
  • methods of the invention have found that single molecule sequencing of DNA or protein biomarkers (via nucleic acid ligands) from urine samples show an increased sensitivity as compared to qPCR-based assays of biomarkers from urine samples.
  • single molecule sequencing of DNA and protein biomarkers in urine has comparable sensitivity as qPCR sequencing of DNA and protein biomarkers from tissue samples, as highlighted in Example 3 below.
  • assays of the invention that detect biomarkers in urine samples have similar performance and sensitivity of invasive tissue-based assays.
  • single molecule sequencing is used to maximize detection of FGFR3 mutations by conducting the biomarker assay on the Ion Torrent PGM platform (Life
  • a primary PCR step is carried out using chimeric primers containing a sequence specific portion for amplifying the exons of interest (Exons 7, 10, and 15) along with adapter sequences required for sequencing analysis.
  • Sequence specific primers suitable for use in smFGFR3 can be designed using any method known in the art. In certain embodiments, the primer can vary in lengths between 16bp to 22 bp.
  • the primary consideration is the Tm of the sequence specific portion. For example, primers with target specific Tm values ranging from ⁇ 52°C to ⁇ 68°C generated successful amplification products with chimeric oligonucleotides.
  • FGFR3 amplicons are designed to be ⁇ 100bp or smaller to accommodate read lengths on the sequencing platform.
  • methods of the invention also provide for single molecule detection of other nucleic acids, such as TWIST1 , VIM, and NID2, and proteins such as MMP-2, MMP-9, and ADAM- 12, through detection of protein-specific nucleic acids.
  • 454 sequencing involves two steps. In the first step, DNA is sheared into fragments of approximately 300-800 base pairs, and the fragments are blunt ended. Oligonucleotide adaptors are then ligated to the ends of the fragments. The adaptors serve as primers for amplification and sequencing of the fragments.
  • the fragments can be attached to DNA capture beads, e.g., streptavidin-coated beads using, e.g., Adaptor B, which contains 5'-biotin tag.
  • the fragments attached to the beads are PCR amplified within droplets of an oil-water emulsion. The result is multiple copies of clonally amplified DNA fragments on each bead.
  • the beads are captured in wells (pico-liter sized). Pyrosequencing is performed on each DNA fragment in parallel. Addition of one or more nucleotides generates a light signal that is recorded by a CCD camera in a sequencing instrument. The signal strength is proportional to the number of nucleotides incorporated. Pyrosequencing makes use of pyrophosphate (PPi) which is released upon nucleotide addition. PPi is converted to ATP by ATP sulfurylase in the presence of adenosine 5' phosphosulfate. Luciferase uses ATP to convert luciferin to oxyluciferin, and this reaction generates light that is detected and analyzed.
  • PPi pyrophosphate
  • SOLiD sequencing genomic DNA is sheared into fragments, and adaptors are attached to the 5' and 3' ends of the fragments to generate a fragment library.
  • internal adaptors can be introduced by ligating adaptors to the 5' and 3' ends of the fragments, circularizing the fragments, digesting the circularized fragment to generate an internal adaptor, and attaching adaptors to the 5' and 3' ends of the resulting fragments to generate a mate-paired library.
  • clonal bead populations are prepared in microreactors containing beads, primers, template, and PCR components.
  • the templates are denatured and beads are enriched to separate the beads with extended templates. Templates on the selected beads are subjected to a 3' modification that permits bonding to a glass slide.
  • the sequence can be determined by sequential hybridization and ligation of partially random oligonucleotides with a central determined base (or pair of bases) that is identified by a specific fluorophore. After a color is recorded, the ligated oligonucleotide is cleaved and removed and the process is then repeated.
  • Illumina sequencing is based on the amplification of DNA on a solid surface using fold-back PCR and anchored primers. Genomic DNA is fragmented, and adapters are added to the 5' and 3' ends of the fragments. DNA fragments that are attached to the surface of flow cell channels are extended and bridge amplified. The fragments become double stranded, and the double stranded molecules are denatured. Multiple cycles of the solid-phase amplification followed by denaturation can create several million clusters of approximately 1 ,000 copies of single-stranded DNA molecules of the same template in each channel of the flow cell.
  • Primers DNA polymerase and four fluorophore-labeled, reversibly terminating nucleotides are used to perform sequential sequencing. After nucleotide incorporation, a laser is used to excite the fluorophores, and an image is captured and the identity of the first base is recorded. The 3' terminators and fluorophores from each incorporated base are removed and the incorporation, detection and identification steps are repeated.
  • SMRT single molecule, real-time
  • each of the four DNA bases is attached to one of four different fluorescent dyes. These dyes are phospholinked.
  • a single DNA polymerase is immobilized with a single molecule of template single stranded DNA at the bottom of a zero-mode waveguide (ZMW).
  • ZMW is a confinement structure which enables observation of incorporation of a single nucleotide by DNA polymerase against the background of fluorescent nucleotides that rapidly diffuse in an out of the ZMW (in microseconds). It takes several milliseconds to incorporate a nucleotide into a growing strand.
  • the fluorescent label is excited and produces a fluorescent signal, and the fluorescent tag is cleaved off. Detection of the corresponding fluorescence of the dye indicates which base was incorporated. The process is repeated.
  • a nanopore is a small hole, of the order of 1 nanometer in diameter. Immersion of a nanopore in a conducting fluid and applicatiorrof a potential across it results in a slight electrical current due to conduction of ions through the nanopore. The amount of current which flows is sensitive to the size of the nanopore. As a DNA molecule passes through a nanopore, each nucleotide on the DNA molecule obstructs the nanopore to a different degree. Thus, the change in the current passing through the nanopore as the DNA molecule passes through the nanopore represents a reading of the DNA sequence.
  • chemFET chemical-sensitive field effect transistor
  • DNA molecules can be placed into reaction chambers, and the template molecules can be hybridized to a sequencing primer bound to a polymerase. Incorporation of one or more triphosphates into a new nucleic acid strand at the 3' end of the sequencing primer can be detected by a change in current by a chemFET.
  • An array can have multiple chemFET sensors.
  • single nucleic acids can be attached to beads, and the nucleic acids can be amplified on the bead, and the individual beads can be transferred to individual reaction chambers on a chemFET array, with each chamber having a chemFET sensor, and the nucleic acids can be sequenced.
  • Another example of a sequencing technique that can be used in the methods of the provided invention involves using an electron microscope (Moudrianakis E. N. and Beer M. Proc Natl Acad Sci USA. 1965 March; 53:564-71).
  • individual DNA molecules are labeled using metallic labels that are distinguishable using an electron microscope. These molecules are then stretched on a flat surface and imaged using an electron microscope to measure sequences.
  • methods of the invention provide for detection of methylation patterns in nucleic acids.
  • Methods include a number of bisulfite treatment sequencing methods in which genomic DNA is isolated and treated with bisulfite.
  • Bisulfite DNA sequencing utilizes bisulfite-induced modification of genomic DNA under conditions whereby unmethylated cytosine is converted to uracil.
  • the bisulfite-modified sequence is then amplified by PCR with two sets of strand-specific primers to yield a pair of fragments, one from each strand, in which all uracil and thymine residues are amplified as thymine and only 5-methylcytosine residues are amplified as cytosine;
  • the PCR products can be sequenced or can be cloned and sequenced to provide methylation maps of single DNA molecules. See Frommer, M. et al., Proc. Natl. Acad. Sci. 89: 1827-1831 (1992).
  • a barcode be ligated to the bisulfite modified targets and the methylated sample library can be pooled with other target nucleic acids and/or nucleic acids for multiplex sequencing.
  • MSP methylation specific PCR
  • hypermethylated and are potential biomarkers.
  • Sequences can be read that originate from a single molecule or that originate from amplifications from a single molecule. Millions of independent amplifications of single molecules can be performed in parallel either on a solid surface or in tiny compartments in water/oil emulsion.
  • the DNA sample to be sequenced can be diluted and/or dispersed sufficiently to obtain one molecule in each compartment. This dilution can be followed by DNA amplification to generate copies of the original DNA sequences and creating "clusters" of molecules all having the same sequence. These clusters can then be sequenced. Many millions of reads can be generated in one run.
  • Sequence can be generated starting at the 5' end of a given strand of an amplified sequence and/or sequence can be generated from starting from the 5' end of the complementary sequence.
  • sequence from strands is generated, i.e. paired end reads (see for example, Harris, U.S. patent number 7,767,400).
  • Nucleotides useful in the invention include any nucleotide or nucleotide analog, whether naturally-occurring or synthetic.
  • preferred nucleotides include phosphate esters of deoxyadenosine, deoxycytidine, deoxyguanosine, deoxythymidine, adenosine, cytidine, guanosine, and uridine.
  • Other nucleotides useful in the invention comprise an adenine, cytosine, guanine, thymine base, a xanthine or hypoxanthine; 5-bromouracil, 2-aminopurine,
  • deoxyinosine or methylated cytosine, such as 5-methylcytosine, and N4-methoxydeoxycytosine.
  • bases of polynucleotide mimetics such as methylated nucleic acids, e.g., 2'-0- methRNA, peptide nucleic acids, modified peptide nucleic acids, locked nucleic acids and any other structural moiety that can act substantially like a nucleotide or base, for example, by exhibiting base-complementarity with one or more bases that occur in DNA or RNA and/or being capable of base-complementary incorporation, and includes chain-terminating analogs.
  • a nucleotide corresponds to a specific nucleotide species if they share base-complementarity with respect to at least one base.
  • Nucleotides for nucleic acid sequencing according to the invention preferably include a detectable label that is directly or indirectly detectable.
  • Preferred labels include optically- detectable labels, such as fluorescent labels.
  • fluorescent labels include, but are not limited to, 4-acetamido-4'-isothiocyanatostilbene-2,2'disulfonic acid; acridine and derivatives: acridine, acridine isothiocyanate; 5-(2'-aminoethyl)aminonaphthalene-l-sulfonic acid (EDANS); 4-amino-N-[3-vinylsulfonyl)phenyl]naphthalimide-3,5 disulfonate; N-(4-anilino-l- naphthyl)maleimide; anthranilamide; BODIPY; Brilliant Yellow; coumarin and derivatives; coumarin, 7-amino-4-methylcoumarin (AMC, Coumarin 120), 7-
  • Nucleic acid polymerases generally useful in the invention include DNA polymerases, RNA polymerases, reverse transcriptases, and mutant or altered forms of any of the foregoing. DNA polymerases and their properties are described in detail in, among other places, DNA Replication 2nd edition, Kornberg and Baker, W. H. Freeman, New York, N.Y. (1991). Known conventional DNA polymerases useful in the invention include, but are not limited to,
  • Pyrococcus furiosus (Pfu) DNA polymerase (Lundberg et al., 1991 , Gene, 108: 1 , Stratagene), Pyrococcus woesei (Pwo) DNA polymerase (Hinnisdaels et al., 1996, Biotechniques, 20: 186-8, Boehringer Mannheim), Thermus thermophilus (Tth) DNA polymerase (Myers and Gelfand 1991 , Biochemistry 30:7661), Bacillus stearothermophilus DNA polymerase (Stenesh and McGowan, 1977, Biochim Biophys Acta 475:32), Thermococcus litoralis (Tli) DNA polymerase (also referred to as Vent.TM.
  • DNA polymerase Cariello et al., 1991, Polynucleotides Res, 19: 4193, New England Biolabs), 9.degree.Nm.TM. DNA polymerase (New England Biolabs), Stoffel fragment, ThermoSequenase® (Amersham Pharmacia Biotech UK), Therminator.TM. (New England Biolabs), Thermotoga maritima (Tma) DNA polymerase (Diaz and Sabino, 1998 Braz J; Med. Res, 31 : 1239), Thermus aquaticus (Taq) DNA polymerase (Chien et al., 1976, J.
  • thermophilic polymerases Both mesophilic polymerases and thermophilic polymerases are contemplated.
  • Thermophilic DNA polymerases include, but are not limited to, ThermoSequenase®,
  • Reverse transcriptases useful in the invention include, but are not limited to, reverse transcriptases from HIV, HTLV-1, HTLV-II, FeLV, FIV, SIV, AMV, MMTV, MoMuLV and other retroviruses (see Levin, Cell 88:5-8 (1997); Verma, Biochim Biophys Acta. 473:1-38 (1977); Wu et al., CRC Crit. Rev Biochem. 3:289-347 (1975)).
  • nucleic acid template molecules are attached to a substrate (also referred to herein as a surface) and subjected to analysis by single molecule sequencing as described herein. Nucleic acid template molecules are attached to the surface such that the template/primer duplexes are individually optically resolvable.
  • Substrates for use in the invention can be two- or three-dimensional and can comprise a planar surface (e.g., a glass slide) or can be shaped.
  • a substrate can include glass (e.g., controlled pore glass (CPG)), quartz, plastic (such as polystyrene (low cross-linked and high cross-linked polystyrene), polycarbonate, polypropylene and poly(methymethacrylate)), acrylic copolymer, polyamide, silicon, metal (e.g., alkanethiolate- derivatized gold), cellulose, nylon, latex, dextran, gel matrix (e.g., silica gel), polyacrolein, or composites.
  • CPG controlled pore glass
  • plastic such as polystyrene (low cross-linked and high cross-linked polystyrene), polycarbonate, polypropylene and poly(methymethacrylate)
  • acrylic copolymer polyamide
  • silicon e.g., metal (e.g., alkanethiolate- derivatized gold)
  • cellulose e.g., nylon, latex, dextran, gel matrix (e.g.
  • Suitable three-dimensional substrates include, for example, spheres, microparticles, beads, membranes, slides, plates, micromachined chips, tubes (e.g., capillary tubes), microwells, microfluidic devices, channels, filters, or any other structure suitable for anchoring a nucleic acid.
  • Substrates can include planar arrays or matrices capable of having regions that include populations of template nucleic acids or primers. Examples include nucleoside-derivatized CPG and polystyrene slides; derivatized magnetic slides; polystyrene grafted with polyethylene glycol, and the like.
  • Substrates are preferably coated to allow optimum optical processing and nucleic acid attachment. Substrates for use in the invention can also be treated to reduce background.
  • Exemplary coatings include epoxides, and derivatized epoxides (e.g., with a binding molecule, such as an oligonucleotide or streptavidin).
  • Various methods can be used to anchor or immobilize the nucleic acid molecule to the surface of the substrate.
  • the immobilization can be achieved through direct or indirect bonding to the surface.
  • the bonding can be by covalent linkage. See, Joos et al., Analytical Biochemistry 247:96-101 , 1997; Oroskar et al., Clin. Chem. 42: 1547-1555, 1996; and handjian, Mol. Bio. Rep. 1 1 : 107-1 15, 1986.
  • a preferred attachment is direct amine bonding of a terminal nucleotide of the template or the 5' end of the primer to an epoxide integrated on the surface.
  • the bonding also can be through non-covalent linkage.
  • biotin-streptavidin (Taylor et al., J. Phys. D. Appl. Phys. 24: 1443, 1991) and digoxigenin with anti-digoxigenin (Smith et al., Science 253: 1 122, 1992) are common tools for anchoring nucleic acids to surfaces and parallels.
  • the attachment can be achieved by anchoring a hydrophobic chain into a lipid monolayer or bilayer.
  • Other methods for known in the art for attaching nucleic acid molecules to substrates also can be used.
  • exemplary detection methods include radioactive detection, optical absorbance detection, e.g., UV-visible absorbance detection, optical emission detection, e.g., fluorescence or chemiluminescence.
  • extended primers can be detected on a substrate by scanning all or portions of each substrate simultaneously or serially, depending on the scanning method used.
  • fluorescence labeling selected regions on a substrate may be serially scanned one-by- one or row-by-row using a fluorescence microscope apparatus, such as described in Fodor (U.S. Pat. No. 5,445,934) and Mathies et al. (U.S. Pat. No. 5,091 ,652).
  • Devices capable of sensing fluorescence from a single molecule include scanning tunneling microscope (siM) and the atomic force microscope (AFM). Hybridization patterns may also be scanned using a CCD camera (e.g., Model TE/CCD512SF, Princeton Instruments, Trenton, N.J.) with suitable optics (Ploem, in Fluorescent and Luminescent Probes for Biological Activity Mason, T. G. Ed., Academic Press, Landon, pp. 1 -1 1 (1993), such as described in Yershov et al., Proc. Natl. Acad. Sci. 93:4913 (1996), or may be imaged by TV monitoring.
  • CCD camera e.g., Model TE/CCD512SF, Princeton Instruments, Trenton, N.J.
  • suitable optics Ploem, in Fluorescent and Luminescent Probes for Biological Activity Mason, T. G. Ed., Academic Press, Landon, pp. 1 -1 1 (1993), such as
  • a phosphorimager device For radioactive signals, a phosphorimager device can be used (Johnston et al., Electrophoresis, 13:566, 1990; Drmanac et al., Electrophoresis, 13:566, 1992; 1993).
  • Other commercial suppliers of imaging instruments include General Scanning Inc., (Watertown, Mass. on the World Wide Web at genscan.com), Genix
  • Optical setups include near-field scanning microscopy, far-field confocal microscopy, wide-field epi-illumination, light scattering, dark field microscopy, photoconversion, single and/or multiphoton excitation, spectral wavelength discrimination, fluorophor identification, evanescent wave illumination, and total internal reflection fluorescence (TIRF) microscopy.
  • TIRF total internal reflection fluorescence
  • certain methods involve detection of laser-activated fluorescence using a microscope equipped with a camera.
  • Suitable photon detection systems include, but are not limited to, photodiodes and intensified CCD cameras.
  • an intensified charge couple device (ICCD) camera can be used.
  • ICCD intensified charge couple device
  • the use of an ICCD camera to image individual fluorescent dye molecules in a fluid near a surface provides numerous advantages.
  • ICCD optical setup it is possible to acquire a sequence of images (movies) of fluorophores.
  • Some embodiments of the present invention use TIRF microscopy for imaging.
  • TIRF microscopy uses totally internally reflected excitation light and is well known in the art. See, e.g., the World Wide Web at nikon-instruments.jp/eng/page/products/tirf.aspx.
  • detection is carried out using evanescent wave illumination and total internal reflection fluorescence microscopy.
  • An evanescent light field can be set up at the surface, for example, to image fluorescently-labeled nucleic acid molecules.
  • a laser beam is totally reflected at the interface between a liquid and a solid substrate (e.g., a glass)
  • the excitation light beam penetrates only a short distance into the liquid.
  • the optical field does not end abruptly at the reflective interface, but its intensity falls off exponentially with distance.
  • This surface electromagnetic field, called the "evanescent wave” can selectively excite fluorescent molecules in the liquid near the interface.
  • the thin evanescent optical field at the interface provides low background and facilitates the detection of single molecules with high signal-to-noise ratio at visible wavelengths.
  • the evanescent field also can image fluorescently-labeled nucleotides upon their incorporation into the attached template/primer complex in the presence of a polymerase. Total internal reflectance fluorescence microscopy is then used to visualize the attached
  • Some embodiments of the invention use non-optical detection methods such as, for example, detection using nanopores (e.g., protein or solid state) through which molecules are individually passed so as to allow identification of the molecules by noting characteristics or changes in various properties or effects such as capacitance or blockage current flow (see, for example, Stoddart et al, Proc. Nat. Acad. Sci., 106:7702, 2009; Purnell and Schmidt, ACS Nano, 3 :2533, 2009; Branton et al, Nature Biotechnology, 26: 1 146, 2008; Polonsky et al, U.S.
  • nanopores e.g., protein or solid state
  • Alignment and/or compilation of sequence results obtained from the image stacks produced as generally described above utilizes look-up tables that take into account possible sequences changes (due, e.g., to errors, mutations, etc.). Essentially, sequencing results obtained as described herein are compared to a look-up type table that contains all possible reference sequences plus 1 or 2 base errors.
  • a plurality of nucleic acid molecules being sequenced is bound to a solid support.
  • a capture sequence/universal priming site can be added at the 3' and/or 5' end of the template.
  • the nucleic acids may be bound to the solid support by hybridizing the capture sequence to a complementary sequence covalently attached to the solid support.
  • the capture sequence (also referred to as a universal capture sequence) is a nucleic acid sequence complimentary to a sequence attached to a solid support that may dually serve as a universal primer.
  • the capture sequence is polyNn, wherein N is U, A, T, G, or C, e g., 20-70, 40-60, e.g., about 50.
  • the capture sequence could be polyT40-50 or its complement.
  • a member of a coupling pair such as, e.g., antibody/antigen, receptor/ligand, or the avidin-biotin pair as described in, e.g., U.S. Patent Application No. 2006/0252077
  • a coupling pair such as, e.g., antibody/antigen, receptor/ligand, or the avidin-biotin pair as described in, e.g., U.S. Patent Application No. 2006/0252077
  • a barcode sequence is attached to the nucleic acid, the nucleic acid, or both. See for example, Steinman et al. (PCT internal application number
  • the present invention relates to a kit comprising a detection reagent which binds to any nucleic acid sequence of ADAM 12, GSTPl , FGFR3, MMP2, TWISTI, NID2, Vimentin, and/or p53, and/or polypeptides encoded thereby for the determination of bladder cancer.
  • kits for screening for, assessing the prognosis of an individual with bladder cancer which comprises a reagent selected from the group consisting of: (a) a reagent for detecting mRNA of the ADAM12, GSTPl, FGFR3, MMP2, TWISTI , NID2, Vimentin, and/or p53 gene; (b) a reagent for detecting protein levels of ADAM 12, GSTPl, FGFR3, MMP2, TWISTI, NID2, Vimentin, and/or p53; and (c) a reagent for detecting the biological activity of the ADAM 12, GSTPl , FGFR3, MMP2, TWISTI, NID2, Vimentin, and/or p53.
  • Kits encompassed by the invention comprise materials and reagents required for measuring the expression of such protein and RNA products.
  • the kits may further comprise one or more additional reagents employed in the various methods, such as: (1) reagents for stabilizing and/or purifying RNA from the sample (2) primers for generating test nucleic acids; (3) dNTPs and/or rNTPs (either premixed or separate), optionally with one or more uniquely labelled dNTPs and/or rNTPs (e.g., biotinylated or Cy3 or Cy5 tagged dNTPs);
  • post synthesis labelling reagents such as chemically active derivatives of fluorescent dyes
  • enzymes such as reverse transcriptases, DNA polymerases, and the like
  • various buffer mediums e.g., reaction, hybridization and washing buffers
  • labelled probe purification reagents and components like spin columns, etc.
  • protein purification reagents e.g., protein purification reagents
  • signal generation and detection reagents e.g., streptavidin-alkaline phosphatase conjugate
  • chemifluorescent or chemiluminescent substrate and the like.
  • kits comprise prelabeled quality controlled protein and or RNA isolated from a sample (e.g., blood or chondrocytes or synovial fluid) for use as a control.
  • a sample e.g., blood or chondrocytes or synovial fluid
  • the kits are RT-PCR or qRT-PCR kits.
  • the kits are nucleic acid arrays and protein arrays.
  • Such kits according to the subject invention will at least comprise an array having associated protein or nucleic acid members of the invention and packaging means therefore.
  • the protein or nucleic acid members of the invention may be pre-packaged onto an array.
  • kits are quantitative RT-PCR kits.
  • the quantitative RT-PCR kit includes the following: (a) primers used to amplify each of a combination of biomarkers of the invention; (b) buffers and enzymes including an reverse transcriptase; (c) one or more thermos table polymerases; and (d) Sybr® Green.
  • the kit of the invention also includes (a) a reference control RNA and (b) a spiked control RNA.
  • kits that are useful for (a) diagnosing individuals as having bladder cancer and/or early stage bladder cancer.
  • the invention also provides kits that are useful for determining the likelihood of bladder cancer in patients presented with hematuria. Additional embodiments of the invention include kits that are useful for monitoring the recurrence of bladder cancer.
  • a kit is comprised a forward and reverse primer wherein the forward and reverse primer are designed to quantitate expression of all of the species of mRNA corresponding to each of the biomarkers as identified in accordance with the invention useful in determining whether an individual has bladder cancer and/or early stage bladder cancer or not.
  • at least one of the primers is designed to span an exon junction.
  • kits that are useful for detecting, diagnosing, monitoring and prognosing bladder cancer based upon the detection of protein or RNA products of AD AMI 2, GSTP1 , FGFR3, MMP2, TWIST1, NID2, Vimentin, and/or p53, possibly in combination with at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, all or any combination of the combinatorial biomarkers of the invention in a sample.
  • kits do not include the materials and reagents for measuring the expression of a protein or RNA product of a biomarker of the invention that has been suggested by the prior art to be associated with bladder cancer.
  • such kits include the materials and reagents for measuring the expression of a protein or RNA product of a combinatorial biomarker of the invention that has been suggested by the prior art to be associated with bladder cancer and at least 1 , at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45 or more genes other than the combinatorial biomarkers of the invention.
  • kits do not include the materials and reagents for measuring the expression of a protein or RNA product of a biomarker of the invention that has been suggested by the prior art to be associated with bladder cancer.
  • such kits include the materials and reagents for measuring the expression of a protein or RNA product of ADAM 12, GSTP1 , FGFR3, MMP2, TWISTI , NID2, Vimentin, and/or p53, possibly in combination with a biomarker that has been suggested by the prior art to be associated with bladder cancer and any number of up to at least 1 , at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45 or more genes other than the combinatorial biomarkers of the invention.
  • kits useful for determining whether a subject will be responsive to a therapy based upon detecting a protein or RNA product of ADAM 12, GSTP1, FGFR3, MMP2, TWISTI , NID2, Vimentin, and/or p53, possibly in combination with any number of up to at least 1 , at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, all or any combination of the combinatorial biomarkers of the invention in a sample.
  • kits comprise materials and reagents that are necessary for measuring the expression of a RNA product of a biomarker of the invention.
  • a kit may comprise a microarray or RT-PCR kit.
  • the kits generally comprise probes attached to a solid support surface. The probes may be labelled with a detectable label.
  • the probes are specific for an exon(s), an intron(s), an exon junction(s), or an exon-intron junction(s)), of RNA products of ADAM12 possibly in combination with any number of up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention.
  • the microarray kits may comprise instructions for performing the assay and methods for interpreting and analyzing the data resulting from the performance of the assay.
  • the kits comprise instructions for diagnosing bladder cancer.
  • the kits may also comprise hybridization reagents and/or reagents necessary for detecting a signal produced when a probe hybridizes to a target nucleic acid sequence.
  • the materials and reagents for the microarray kits are in one or more containers. Each component of the kit is generally in its own a suitable container.
  • kits generally comprise pre-selected primers specific for particular RNA products (e.g., an exon(s), an intron(s), an exon junction(s), and an exon-intron junction(s)) of ADAM 12, GSTPl, FGFR3, MMP2, TWISTl , NID2, Vimentin, and/or p53 possibly in combination with any number of up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention.
  • RNA products e.g., an exon(s), an intron(s), an exon junction(s), and an exon-intron junction(s)
  • ADAM 12 e.g., an exon(s), an intron(s), an exon junction(s), and an exon-intron junction(s)
  • ADAM 12 e.g., an exon(s), an intron(s), an exon junction(s), and an exon-intron
  • the RT-PCR kits may also comprise enzymes suitable for reverse transcribing and/or amplifying nucleic acids (e.g., polymerases such as Taq), and deoxynucleotides and buffers needed for the reaction mixture for reverse transcription and amplification.
  • the RT-PCR kits may also comprise probes specific for RNA products of ADAM 12, GSTPl , FGFR3, MMP2, TWISTl , NID2, VIMENTIN, and/or p53, and possibly any number of up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention.
  • the probes may or may not be labelled with a detectable label (e.g., a fluorescent label).
  • kits generally comprise distinct containers suitable for each individual reagent, enzyme, primer and probe.
  • the RT-PCR kits may comprise instructions for performing the assay and methods for interpreting and analyzing the data resulting from the performance of the assay.
  • the kits contain instructions for diagnosing bladder cancer.
  • the kit is a real-time RT-PCR kit.
  • a kit may comprise a 96 well plate and reagents and materials necessary for e.g. SYBR Green detection.
  • the kit may comprise reagents and materials so that beta-actin can be used to normalize the results.
  • the kit may also comprise controls such as water, phosphate buffered saline, and phage MS2 RNA.
  • the kit may comprise instructions for performing the assay and methods for interpreting and analyzing the date resulting from the performance of the assay.
  • the instructions state that the level of a RNA product of ADAM 12, GSTPl , FGFR3, MMP2, TWISTl , NID2, Vimentin, and/or p53, and possibly any number of up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention should be examined at two concentrations that differ by, e.g., 5 fold to 10-fold.
  • the kit can comprise, for example: (1) a first antibody (which may or may not be attached to a solid support) which binds to ADAM 12, GSTPl , FGFR3, MMP2, TWIST1 , NID2, Vimentin, and/or p53 and any combinatorial protein of interest (e.g., a protein product of any number of up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention); and, optionally, (2) a second, different antibody which binds to either the protein, or the first antibody and is conjugated to a detectable label (e.g., a fluorescent label, radioactive isotope or enzyme).
  • a detectable label e.g., a fluorescent label, radioactive isotope or enzyme
  • the antibody-based kits may also comprise beads for conducting an immunoprecipitation. Each component of the antibody-based kits is generally in its own suitable container/Thus, these kits generally comprise distinct containers suitable for each antibody. Further, the antibody-based kits may comprise instructions for performing the assay and methods for interpreting and analyzing the data resulting from the performance of the assay.
  • kits contain instructions for diagnosing bladder cancer.
  • EXAMPLE 1 Whole urine DNA recovery versus DNA recovery from cellular and cell-free portions of whole urine.
  • the amount of DNA recoverable from a whole urine sample was compared to the amount of DNA recoverable from a cellular (pellet) urine sample and a cell-free (supernantant) urine sample from an identical sample of whole urine.
  • the second 50ml conical is labeled "Pellet” (Cell-associated DNA) and is centrifuged at 1800g for 10 minutes. The supernatant is poured off into a new 50ml conical and is labeled "Free DNA”.
  • Pellet Cell-associated DNA
  • the amount of DNA required to detect 0.08% mutant DNA with 85% confidence is 22ng.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Hospice & Palliative Care (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Oncology (AREA)
  • Biotechnology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention provides methods for detecting disease, e.g. cancer, using a combination of nucleic acid biomarkers in both the cellular and the cell-free portion of a body fluid sample. In particular, the described methods can be used to analyze the nucleic acids in whole urine to determine whether a subject has cancer, e.g., prostate or bladder cancer.

Description

DIAGNOSTIC ASSAY COMBINING CELLULAR AND CELL FREE NUCLEIC ACID
FIELD OF THE INVENTION
The invention generally relates to methods for screening a body fluid for markers of disease.
BACKGROUND
Advanced nucleic acid screening techniques have resulted in a revolution in disease screening. It is now possible to screen a patient for a wide variety of markers indicative of disease, such as cancer, birth defects, autoimmune disease, infection, etc.
In order to screen for a disorder, a sample is taken from the subject and prepared to recover nucleic acids of interest, typically DNA. The samples may be cells, such as cells from a biopsy, or the samples may be fluids, such as serum from the blood. When a mixed sample, such as whole blood, is sampled for an assay, either the cellular component (e.g., leukocytes, circulating cells) are precipitated, lysed, and sampled, or the fluid component (e.g., serum) is sampled. Each portion gives different information about the subject.
The standard methods for isolating nucleic acids from a mixed sample present two complications. The first is that extra preparative steps must be taken to isolate the cellular and non-cellular components. These extra steps take time, and increase the risk that the sample may be contaminated or damaged. The second is that a valuable opportunity to learn more about the sample is lost when the other portion of the mixed sample is discarded.
SUMMARY
The present invention provides methods for detecting disease, e.g. cancer, using a combination of nucleic acid biomarkers in both the cellular and the cell-free portion of a body fluid sample. In particular, the described methods can be used to analyze the nucleic acids in whole urine to determine whether a subject has cancer, e.g., prostate or bladder cancer. In some instances, the two portions of urine are separated and analyzed individually to give a unique insight into the condition of the subject. In some instances, the condition of the subject could not have been determined by analyzing the cellular or the cell-free component, alone. In other instances, all of the nucleic acids in the body fluid sample are analyzed without separating the body sample into component parts. Accordingly, the need for separation is reduced while the predictive value of the assay is advantageously increased.
When used with body fluids that can be recovered non-invasively, the techniques make it easier to monitor the progression of a disease without disruptive procedures, such as biopsy. In some aspects of the invention, the method includes collecting a urine sample and analyzing it to detect DNA biomarkers associated with bladder cancer or prostate cancer. The technique can also be used to stratify patients based on their likelihood of disease. In some aspects of the invention, the biomarkers detected include FGFR3, MMP2, TWIST1 , Vimentin and NID2. In some embodiments, epigenetic changes, such as methylation of nucleic acids, can be evaluated in order to determine the condition of a subject. In other embodiments, biomarkers can be detected with single-molecule sequencing. The results of these tests are then compared to established reference ranges and are evaluated in combination to determine the likelihood of cancer.
The combination of biomarkers contemplated by the invention is able to provide both high positive and negative predictive values across all stages and grades of cancer, e.g., bladder cancer or prostate cancer. In accordance with certain aspects of the invention, two marker cutoffs are established; one cutoff to maximize sensitivity and negative predictive value and a second cutoff to maximize specificity and positive predictive value. Marker thresholds are then set to provide maximum NPV and sensitivity, such that patients who do not have cancer might be excluded from further intervention. By setting marker cutoffs to high PPV and specificity, patients could also be triaged into those that might benefit from maximum intervention. Patients assessed in between those cutoffs might continue to receive standard intervention. In more specific aspects of the invention, patients with all biomarkers below a predetermined cutoff are determined as having a low likelihood of cancer.
Further aspects and features of the invention will be apparent upon inspection of the following detailed description thereof.
DETAILED DESCRIPTION
Methods of the invention provide a sensitive and specific test for detecting and diagnosing different diseases or disorders, particularly cancer. In certain aspects, the screening assay includes identifying a nucleic acid sample associated with a cellular component of a body fluid as well as a nucleic acid that is not associated with a cellular component, e.g, "cell-free" or "circulating" nucleic acids. In some instances, the measurements are compared to threshold parameters indicative of the absence of cancer.
The biomarkers chosen are immaterial to the operation of the invention as long as the marker is associated with the disease for which screening is being conducted. Exemplary biomarkers include nucleic acid biomarkers for cancer, infectious diseases, autoimmune diseases, and birth defects. Biomarkers used in methods of the invention are chosen based upon their predictive value or suspected predictive value for the condition or conditions being diagnosed. Particular markers are selected based upon various diagnostic criteria, such as suspected association with disease. The number of markers chosen will depend on the number of assays performed and is at the discretion of the user. Biomarkers should be chosen that cumulatively increase the specificity/sensitivity of the assay. A panel of markers can be chosen to increase the effectiveness of diagnosis, prognosis, treatment response, and/or recurrence. In addition to general concerns around specificity and sensitivity, markers can also be chosen in consideration of the patient's history and lifestyle. For example, other diseases that the patient has, might have, or has had can effect the choice of the panel of biomarkers to be analyzed. Drugs that the patient has in his/her system may also affect biomarker selection.
Threshold values for any particular biomarker and associated disease are determined by reference to literature or standard of care criteria or may be determined empirically. In certain embodiments of the invention, thresholds for use in association with biomarkers of the invention are based upon positive and negative predictive values associated with threshold levels of the marker. There are numerous methods for determining thresholds for use in the invention, including reference to standard values in the literature or associated standards of care. The precise thresholds chosen are immaterial as long as they have the desired association with diagnostic output.
The invention is applicable to diagnosis and monitoring of any disease, either in symptomatic or asymptomatic patient populations. For example, the invention can be used for diagnosis of infectious diseases, inherited diseases, and other conditions, such as disease or damage caused by drug or alcohol abuse. The invention can also be applied to assess therapeutic efficacy, potential for disease recurrence or spread (e.g. metastasis).
Methods of the invention can be used on patients known to have a disease, or can be used to screen healthy subjects on a periodic basis. Screening can be done on a regular basis (e.g., weekly, monthly, annually, or other time interval); or as a one-time event. The outcome of the analysis may be used to alter the frequency and/or type of screening, diagnostic and/or treatment protocols. Different conditions can be screened for at different time intervals and as a function of different risk factors (e.g., age, weight, gender, history of smoking, family history, genetic risks, exposure to toxins and/or carcinogens etc., or a combination thereof). The particular screening regimen and choice of markers used in connection with the invention are determined at the discretion of the physician or technician.
Biomarkers associated with diseases are shown for example in Shuber (U.S. patent application number 2009/0075266), the content of which is incorporated by reference herein in its entirety. The invention is especially useful in screening for cancer. Examples of biomarkers associated with cancer include FGFR3, matrix metalloproteinase (MMP), neutrophil gelatinase- associated lipocalin (NGAL), MMP/NGAL complex, thymosin β15, thymosin β16, collagen like gene (CLG) product, prohibitin, glutathione-S-transferase, beta-5-tubulin, ubiquitin,
tropomyosin, Cyr61, cystatin B, chaperonin 10, and profilin. Examples of MMPs include, but are not limited to, MMP-2, MMP-9, MMP9/ GAL complex, MMP/TIMP complex, MMP/TIMP1 complex, ADAMTS-7 or ADAM- 12, among others.
Biomarkers associated with development of breast cancer are shown in Eriander et al. (US 7,504,214), Dai et al. (US 7,514, 209 and 7, 171 ,31 1), Baker et al. (US 7,056,674 and US 7,081 ,340), Eriander et al. (US 2009/0092973). The contents of the patent application and each of these patents are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with breast cancer include: ErbB2 (Her2); ESR1 ; BRCA1 ; BRCA2; p53; mdm2; cyclinl ; p27; B_Catenin; BAG 1 ; B1N1 ; BUB 1 ; C20_orfl ; CCNB 1 ; CCNE2;
CDC20; CDH1 ; CEGP1 ; CIAP1 ; cMYC; CTSL2; DKFZp586M07; DR5; EpCAM; EstRl ; FOXM1 ; GRB7; GSTM1 ; GSTM3; HER2; HNRPAB; ID 1 ; IGF 1 R; ITGA7; Ki_67; KNSL2; LMNB 1; MCM2; MELK; MMP 12; MMP9; MYBL2; NEK2; NME1 ; NPD009; PCNA; PR; PREP; PTTG1 ; RPLPO; Src; STK15; STMY3; SURV; TFRC; TOP2A; and TS.
Biomarkers associated with development of cervical cancer are shown in Patel (US 7,300,765), Pardee et al. (US 7, 153,700), Kim (US 6,905,844), Roberts et al. (US 6,316,208), Schlegel (US 2008/01 13340), Kwok et al. (US 2008/0044828), Fisher et al. (US 2005/0260566), Sastry et al. (US 2005/0048467), Lai (US 2008/03 1 1570) and Van Der Zee et al. (US
2009/0023137). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with cervical cancer include: SC6; SIX 1 ; human cervical cancer 2 protooncogene (HCCR-2); p27; virus oncogene E6; virus oncogene E7; ρ16ΓΝ 4Α; Mem proteins (such as Mcm5); Cdc proteins; topoisomerase 2 alpha; PCNA; Ki-67; Cyclin E; p-53; PAll ; DAP-kinase; ESR1 ; APC; TIMP-3; RAR-β; CALCA; TSLC1 ; TIMP-2; DcRl ; CUDR; DcR2; BRCA1 ; pl5; MSH2; RassflA; MLH1 ; MGMT; SOX1 ; PAX1 ; LMX1A; NKX6-1 ; WT1 ; ONECUT1 ;
SPAG9; and Rb (retinoblastoma) proteins.
Biomarkers associated with development of vaginal cancer are shown in Giordano (US 5,840,506), Kruk (US 2008/0009005), Hellman et al. (Br J Cancer. 100(8): 1303-1314, 2009). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with vaginal cancer include: pRb2/pl 30 and Bcl-2.
Biomarkers associated with development of brain cancers (e.g., glioma, cerebellum, medulloblastoma, astrocytoma, ependymoma, glioblastoma) are shown in D'Andrea (US 2009/0081237), Murphy et al. (US 2006/0269558), Gibson et al. (US 2006/0281089), and Zetter et al. (US 2006/0160762). The contents of each of the articles and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with brain cancers include: epidermal growth factor receptor (EGFR);
phosphorylated PKB/Akt; EGFRvIIl; FANCI; Nr-CAM; antizyme inhibitor (AZI); BNIP3; and miRNA-2 .
Biomarkers associated with development of renal cancer are shown in Patel (US
7,300,765), Soyupak et al. (US 7,482, 129), Sahin et al. (US 7,527,933), Price et al. (US
7,229,770), Raitano (US 7,507,541 ), and Becker et al. (US 2007/0292869). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with renal cancers include: SC6; 36P6D5; IMP3; serum amyloid alpha; YKL-40; SC6; and carbonic anhydrase IX (CA IX).
Biomarkers associated with development of hepatic cancers (e.g., hepatocellular carcinoma) are shown in Home et al. (US 6,974,667), Yuan et al. (US 6,897,018), Hanausek- Walaszek et al. (US 5,310,653), and Liew et al. (US 2005/0152908). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with hepatic cancers include: Tetraspan NET-6 protein; collagen, type V, alpha; glypican 3; pituitary tumor-transforming gene 1 (PTTGl); Galectin 3; solute carrier family 2, member 3, or glucose transporter 3 (GLUT3); metallothionein 1 L; CYP2A6; claudin 4; serine protease inhibitor, Kazal type I (SPINK.1); DLC-1 ; AFP; HSP70; CAP2; glypican 3; glutamine synthetase; AFP; AST and CEA.
Biomarkers associated with development of gastric, gastrointestinal, and/or esophageal cancers are shown in Chang et al. (US 7,507,532), Bae et al. (US 7,368,255), Muramatsu et al. (US 7,090,983), Sahin et al. (US 7,527,933), Chow et al. (US 2008/0138806), Waldman et al. (US 2005/0100895), Goldenring (US 2008/0057514), An et al. (US 2007/0259368), Guilford et al. (US 2007/0184439), Wirtz et al. (US 2004/0018525), Filella et al. (Acta Oncol. 33(7):747- 751 , 1994), Waldman et al. (US 6,767,704), and Lipkin et al. (Cancer Research, 48:235-245, 1988). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with gastric, gastrointestinal, and/or esophageal cancers include: MH15 (HnlL); RUNX3; midkine;
Chromogranin A (CHGA); Thy-1 cell surface antigen (THY1); IPO-38; CEA; CA 19.9; GroES; TAG-72; TGM3; HE4; LGALS3; ILI RN; TRIP 13; FIGNLl ; CRIPI ; S100A4; EXOSC8; EXPI; CRCA-1 ; BRRN1 ; NELF; EREG; TMEM40; TMEM109; and guanylin cyclase C.
Biomarkers associated with development of ovarian cancer are shown in Podust et al. (US 7,510,842), Wang (US 7,348,142), O'Brien et al. (US 7,291 ,462, 6,942,978, 6,316,213, 6,294,344, and 6,268,165), Ganetta (US 7,078, 180), Malinowski et al. (US 2009/0087849), Beyer et al. (US 2009/0081685), Fischer et al. (US 2009/0075307), Mansfield et al. (US
2009/0004687), Livingston et al. (US 2008/0286199), Farias-Eisner et al. (US 2008/0038754), Ahmed et al. (US 2007/0053896), Giordano (US 5,840,506), and Tchagang et al. (Mol Cancer Ther, 7:27-37, 2008). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with ovarian cancer include: hepcidin; tumor antigen-derived gene (TADG-15); TADG- 12; TADG-14; ZEB; PUMP- 1 ; stratum corneum chymotrytic enzyme (SCCE); NES-1 ; μΡΑ; PAI-2; cathepsin B; cathepsin L; ERCC5; MMP-2; pRb2/pl 30 gene; matrix
metalloproteinase-7 (MMP-7); progesterone-associated endometrial protein (PALP) ; cancer antigen 125 (CA125) ; CTAP3; human epididymis 4 (HL4); plasminogen activator urokinase receptor (PLAUR); MUC-1 ; FGF-2; cSHMT; Tbx3; utrophin; SLPI; osteopontin (SSP1 );
mesothelin (MSLN); SPON 1 ; interleukin-7; folate receptor 1 ; and claudin 3. Biomarkers associated with development of head-and-neck and thyroid cancers are shown in Sidransky et al. (US 7,378,233), Skolnick et al. (US 5,989,815), Budiman et al. (US 2009/0075265), Hasina et al. (Cancer Research, 63 :555-559, 2003), ebebew et al. (US
2008/0280302), and Ralhan (Mol Cell Proteomics, 7(6): 1 162-1 173, 2008). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with head-and-neck and thyroid cancers include: BRAF; Multiple Tumor Suppressor (MTS); PAI-2; stratifin; YWHAZ; S100- A2; S 100-A7 (psoriasin); S100-A1 1 (calgizarrin); prothymosin alpha (PTHA); L-lactate dehydrogenase A chain; glutathione S-transferase Pi; APC-binding protein EB1 ; fascin;
peroxiredoxin2; carbonic anhydrase I; flavin reductase; histone H3; ECM 1 ; TMPRSS4;
ANGPT2; T1MP1; LOXL4; p53; IL-6; EGFR; Ku70; GST-pi; and polybromo- l D.
Biomarkers associated with development of colorectal cancers are shown in Raitano et al. (US 7,507,541), Reinhard et al. (US 7,501 ,244), Waldman et al. (US 7,479,376); Schleyer et al. (US 7,198,899); Reed (US 7, 163,801), Robbins et al. (US 7,022,472), Mack et al. (US
6,682,890), Tahiti et al. (US 5,888,746), Budiman et al. (US 2009/0098542), Karl (US
2009/007531 1 ), Arjol et al. (US 2008/0286801), Lee et al. (US 2008/0206756), Mori et al. (US 2008/0081333), Wang et al. (US 2008/0058432), Belacel et al. (US 2008/0050723), Stedronsky et al. (US 2008/0020940), An et al. (US 2006/0234254), Eveleigh et al. (US 2004/0146921), and Yeatman et al. (US 2006/0195269). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with colorectal cancers include: 36P6D5; TTK; CDX2; NRG4; TUCAN; hMLHl ; hMSH2; M2-PK; CGA7; CJA8; PTP.alpha.; APC; p53; Ki-ras; complement C3a des- arg; alphal -antitrypsin; transferrin; MMP-1 1 ; CA-19-9; TPA; TPS; TIMP-1 ; C lOorfi;
carcinoembryonic antigen (CEA); a soluble fragment of cytokeratin 19 (CYFRA 21- 1 ); TAC1 ; carbohydrate antigen 724 (CA72-4); nicotinamide N-methyltransferase (NNMT); pyrroline-5- carboxylate reductase (PROC); S-adenosylhomocysteine hydrolase (SAHH); 1BABP-L polypeptide; and Septin 9.
Biomarkers associated with development of prostate cancer are shown in Sidransky (US 7,524,633), Platica (US 7,510,707), Salceda et al. (US 7,432,064 and US 7,364,862), Siegler et al. (US 7,361 ,474), Wang (US 7,348, 142), Ali et al. (US 7,326,529), Price et al. (US 7,229,770), O'Brien et al. (US 7,291 ,462), Golub et al. (US 6,949,342), Ogden et al. (US 6,841,350), An et al. (US 6, 171 ,796), Bergan et al. (US 2009/0124569), Bhowmick (US 2009/0017463), Srivastava et al. (US 2008/0269157), Chinnaiyan et al. (US 2008/0222741 ), Thaxton et al. (US
2008/0181850), Dahary et al. (US 2008/0014590), Diamandis et al. (US 2006/0269971), Rubin et al. (US 2006/0234259), Einstein et al. (US 2006/01 15821), Paris et al. (US 2006/01 10759), Condon-Cardo (US 2004/0053247), and Ritchie et al. (US 2009/0127454). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with prostate cancer include: PSA; GSTP1 ; PAR; CSG; M1F; TADG-15; p53; Y L-40; ZEB; HOXC6; Pax 2; prostate-specific transglutaminase; cytokeratin 15; MEK4; MIPl-β; fractalkine; IL- 15; ERG8; EZH2; EPC1 ; EPC2; NLGN-4Y; kallikrein 1 1 ; ABP280 (FLNA); AMACR; AR; BM28; BUB3; CaMKK; CASPASE3; CD 7; DYNAMIN; E2F1 ; E-CADHERI ; EXPORT1N; EZH2; FAS; GAS7; GS28; ICBP90; ITGA5; JAGGED 1 ; JAM1 ; ANADAPTIN; LF6; KRIP1 ; LAP2; MCAM; IB 1 (MKI67); MTA1 ; MUC1 ; MYOSIN- VI; P27; P63; P27; PAXILLIN; PLCLN;
PSA(KL 3); RAB27; RBBP; RI 1 ; SAPKa; TPD52; XIAP; ZAG; and semenogelin II.
Biomarkers associated with development of pancreatic cancer are shown in Sahin et al. (US 7,527,933), Rataino et al. (US 7,507,541), Schleyer et al. (US 7,476,506), Domon et al. (US 7,473,531), McCaffey et al. (US 7,358,231), Price et al. (US 7,229,770), Chan et al. (US 2005/009561 1), Mitchl et al. (US 2006/0258841), and Faca et al. (PLoS Med 5(6):el23, 2008). The contents of each of the articles, patents, and patent applications are incorpo rated by reference herein in their entirety. Exemplary biomarkers that have been associated with pancreatic cancer include: CA19.9; 36P6D5; NRG4; ASCT2; CCR7; 3C4-Ag; KLK1 1 ;
Fibrinogen γ; and Y L40.
Biomarkers associated with development of lung cancer are shown in Sahin et al. (US 7,527,933), Hutteman (US 7,473,530), Bae et al. (US 7,368,255), Wang (US 7,348, 142), Nacht et al. (US 7,332,590), Gure et al. (US 7,314,721), Patel (US 7,300,765), Price et al. (US
7,229,770), O'Brien et al. (US 7,291 ,462 and US 6,316,213), Muramatsu et al. (US 7,090,983), Carson et al. (US 6,576,420), Giordano (US 5,840,506), Guo (US 2009/0062144), Tsao et al. (US 2008/0176236), Nakamura et al. (US 2008/0050378), Raponi et al. (US 2006/0252057), Yip et al. (US 2006/0223127), Pollock et al. (US 2006/0046257), Moon et al. (US 2003/0224509), and Budiman et al. (US 2009/0098543). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with lung cancer include: COX-2; COX4-2; RUNX3; aldoketoreductase family 1 , member B 10; peroxiredoxin 1 (PRDX1); T F receptor superfamily member 18; small proline-rich protein 3 (SPRR3); SOX1 ; SC6; TADG- 15; YKL40; midkine; DAP-kinase;
HOXA9; SCCE; STX1A; HIF1A; CCT3; HLA-DPB1 ; MAFK; RNF5; KIF1 1 ; GHSRl b;
NTSRl ; FOXM1 ; and PUMP-1.
Biomarkers associated with development of skin cancer (e.g., basal cell carcinoma, squamous cell carcinoma, and melanoma) are shown in Roberts et al. (US 6,316,208), Polsky (US 7,442,507), Price et al. (US 7,229,770), Genetta (US 7,078, 180), Carson et al. (US
6,576,420), Moses et al. (US 2008/028681 1), Moses et al. (US 2008/0268473), Dooley et al. (US 2003/0232356), Chang et al. (US 2008/0274908), Alani et al. (US 2008/01 18462), Wang (US 2007/0154889), and Zetter et al. (US 2008/0064047). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with skin cancer include: p27; Cyr61 ; ADAMTS-7;
Cystatin B; Chaperonin 10; Profilin; BRAF; YKL-40; DDX48; erbB3 -binding protein; biliverdin reductase; PLAB; LICAM; SAA; CRP; SOX9; MMP2; CD 10; and ZEB.
Biomarkers associated with development of multiple myeloma are shown in Coignet (US 7,449,303), Shaughnessy et al. (US 7,308,364), Seshi (US 7,049,072), and Shaughnessy et al. (US 2008/0293578, US 2008/0234139, and US 2008/0234138). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with multiple myeloma include: JAG2;
CCND1 ; MAF; MAFB; MMSET; CST6; RAB7L1 ; MAP4K3; HRASLS2; TRAIL; IG; FGL2; GNG1 1 ; MCM2; FLJ10709; TRIM13; NADSYN1 ; TRIM22; AGRN; CENTD2; SESN1 ;
TM7SF2; NICKAPl ; COPG; STAT3; ALOX5; APP; ABCB9; GAA; CEP55; BRCA1 ; ANLN; PYGL; CCNE2; ASPM; SUV39H2; CDC25A; IFIT5; ANKRA2; PHLDB l ; TUBA IA; CDCA7; CDCA2; HFE; RIF 1 ; NEIL3; SLC4A7; FXYD5; MCC; MKNK2; KLHL24; DLC1 ; OPN3; B3GALNT1 ; SPRED1 ; ARHGAP25; RTN2; WNT16; DEPDC1 ; STT3B; ECHDC2; ENPP4; SAT2; SLAMF7; MAN1 C 1 ; INTS7; Z F600; L3MBTL4; LAPTM4B; OSBPL10; KCNS3; THEX1. CYB5D2; UNC93B1; SIDT1 ; TMEM57; HIGD24; FKSG44; C14orf28; LOC387763; TncRNA; C18orfl ; DCU 1 D4; FANCI; ZMAT3; NOTCH1 ; BTG2; RAB1A; TNFRSF10B; HDLBP; R1T1 ; KIF2C; S 100A4; MEIS 1 ; SGOL2; CD302; COX2; C5orf34; FAM1 1 1B;
C 18orf54; and TP53. Biomarkers associated with development of leukemia are shown in Ando et al. (US 7,479,371), Coignet (US 7,479,370 and US 7,449,303), Davi et al. (US 7,416,851), Chiorazzi (US 7,316,906), Seshi (US 7,049,072), Van Baren et al. (US 6, 130,052), Taniguchi (US 5,643,729), Insel et al. (US 2009/0131353), and Van Bockstaele et al. (Blood Rev. 23(l):25-47, 2009). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with leukemia include: SCGF; JAG2; LPL; ADAM29; PDE; Cryptochrome-1 ; CD49d; ZAP-70; PRAME; WT1 ; CD15; CD33; and CD38.
Biomarkers associated with development of lymphoma are shown in Ando et al. (US 7,479,371), Levy et al. (US 7,332,280), and Arnold (US 5,858,655). The contents of each of the articles, patents, and patent applications are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with lymphoma include: SCGF; L 02; BCL6; FN1 ; CCND2; SCYA3; BCL2; CD79a; CD7; CD25; CD45RO; CD45RA; and PRAD1 cyclin.
Biomarkers associated with development of bladder cancer are shown in Price et al. (US 7,229,770), Orntoft (US 6,936,417), Haak-Frendscho et al. (US 6,008,003), Feinstein et al. (US 6,998,232), Elting et al. (US 2008/031 1604), and Wewer et al. (2009/0029372). The contents of each of the patent applications and each of these patents are incorporated by reference herein in their entirety. Exemplary biomarkers that have been associated with bladder cancer include: FGFR3, NT-3; NGF; GDNF; Y L-40; p53; pRB; p21 ; p27; cyclin El ; i67; Fas; urothelial carcinoma-associated 1 ; human chorionic gonadotropin beta type II; insulin-like growth factor- binding protein 7; sorting nexin 16; chondroitin sulfate proteoglycan 6; cathepsin D;
chromodomain helicase DNA-binding protein 2; nell-like 2; tumor necrosis factor receptor superfamily member 7; cytokeratin 18 (C 18); ADAM8; ADAM10; ADAM12; Matrix Metalloproteinase-2 (MMP-2); MMP-9; KAI1 ; and bladder tumor fibronectin (BTF).
In certain circumstances, nucleic acids and proteins associated with a certain cancer vary with respect to the genetic, biochemical, or molecular alterations that associate the nucleic acid or protein with cancer. For example, the cancer causing alterations can include abnormal protein expressions, sequence mutations, methylation patterns, and loss of heterozygosity. Because multiple alterations can be linked to cancer, methods of the invention realize that there is great clinical value in assaying for multiple genetic characteristics across the plurality of biomarkers. In certain aspects, the invention involves obtaining a urine or tissue sample, conducting an assay on the urine or tissue sample to look for a nucleic acid mutation, loss of heterozygosity, and an abnormal protein level, and determining whether the sample is positive or negative for cancer based on the assay. By detecting different alterations in a signal assay, the result is a multimodal analysis that has greater sensitivity and specificity with regard to the diagnosis and
characterization of the disease.
Methods of the invention provide for conducting an assay on a plurality of biomarkers to look for characteristics such as a nucleic acid mutation, a loss of heterozygosity, an abnormal protein level, gene expression patterns, an abnormal methylation pattern, and any other characteristic indicative of cancer. The presence or absence of one or more characteristic is indicative of a positive result for the cancer to be diagnosed. In certain embodiments, the type of characteristic looked for in the plurality of biomarkers is based on the cancer being diagnosed. For example, characteristics associated with bladder cancer include nucleic acid mutations, loss of heterozygosity, abnormal protein levels, and hypermethylation, whereas other cancer types might only be associated with abnormal protein level and hypermethylation patterns. Below the type of characteristics in proteins and nucleic acids that are suitable for use in methods of the invention are exemplified.
Nucleic acid biomarkers are often associated with nucleic acid mutations, which include additions, deletions, insertions, rearrangements, inversions, transitions, transversions, frameshift mutations, nonsense mutations, missense mutations, single nucleotide polymorphisms (SNP) and substitutions of two or more nucleotides within a sequence but not to the extent of large chromosomal sequence changes. SNPs are a type of genomic subtle sequence change that occurs when a single nucleotide replaces another within the sequence. Alterations in chromosome numbers include additions, deletions, inversions, translocations, copy number variations, and substitutions of chromosomes within a sequence. These nucleic acid mutations in biomarkers are often linked to cancer. For example, mutations of the FGFR3 gene and the p53 gene have been observed in bladder cancer. Cappellen D, De Oliveira C, Ricol D, et al., "Frequent activating mutations of FGFR3 in human bladder and cervix carcinomas." NatGenet. 1999;23(1): 18-20; Berggren et al., "p53 mutations in urinary bladder cancer" British Journal of Cancer (2001) 84, 1505-151 1. doi: 10.1054/bjoc.2001.1823. Loss of heterozygosity (LOH) is a common occurrence in patients with cancer. LOH indicates the absence of a functional tumor suppressor gene in the lost region. Loss of heterozygosity results from a deletion or other mutational event within a normal allele at a particular locus heterozygous for a deleterious mutant allele and the normal allele. The mutation in the normal allele renders the cell either hemizygous (one deleterious allele and one deleted allele) or homozygous for the deleterious allele. In other words, the loss of the normal allele is the LOH and may be a genetic determinant in the development of cancer. For example, loss of heterozygosity in the p53 gene is associated with bladder cancer. See Oka et al., "Detection of loss of heterozygosity in the p53 gene in renal cell carcinoma and bladder cancer using the polymerase chain reaction." Molecular Carcinogenesis: Volume 4, Issue 1 , 2006.
In certain embodiments, the level of protein biomarkers in the sample is analyzed in the multi-analyte screening assay to determine if there is an abnormal protein level in the sample. Protein biomarkers are generally considered quantitative biomarkers for which a level or amount of the biomarker present in comparison to a reference level or amount indicates a clinical status. For example, matrix metalloproteinases, such as MMP-2, MMP-9, and metalloproteases, such as ADAM- 12, are associated with bladder cancer. MMPs have been shown to be key regulators of tumor growth, angiogenesis and metastasis formation. Increased MMP expression is required for tumors to grown into the surrounding tissue and for dissemination of metastatic cells into the vasculature and distant sites. Detection of MMPs in the urine of cancer patients has been shown to correlate with disease status in a variety of cancers, including bladder cancer. Biologically active MMP-2 and MMP-9 are found at higher levels and at greater frequency in urine of cancer patients than in healthy controls. In addition, ADAM 12 is expressed in higher levels in cancer subjects than in healthy controls and is described in commonly-owned U.S. Application No. 12/120,544.
In a particular embodiment, methods of the invention optionally include screening for the presence or absence of a methylation pattern in nucleic acid biomarkers, which includes screening nucleic acids for de-methylation, methylation, hypomethylation and hypermethylation. DNA methylation is an important regulator of gene transcription and a large body of evidence has demonstrated that aberrant DNA methylation is associated with unscheduled gene silencing, and the genes with high levels of 5-methylcytosine in their promoter region are transcriptionally silent. Aberrant DNA methylation patterns have been associated with a large number of human malignancies and found in two distinct forms: hypermethylation and hypomethylation compared to normal tissue. Hypermethylation is one of the major epigenetic modifications that repress transcription via promoter region of tumor suppressor genes. Hypermethylation typically occurs at CpG islands in the promoter region and is associated with gene inactivation. Global hypomethylation has also been shown to be causally related to the development and progression of cancer through different mechanisms. For example, a hypermethylation pattern of TWIST1, NID2, and vimentin detected in urine samples is indicative of a positive result for bladder cancer. See Renard I et al., Eur Urol. 2010; 58(1):96-104.
In another embodiment, the multi-analyte screening assay includes screening for gene expression of nucleic acids. Nucleic acid biomarkers associated with gene expression are generally considered quantitative biomarkers for which a level or amount of the biomarker present in comparison to a reference level or amount indicates a clinical status. For example, genes that exhibited significant over-expression in bladder cancer v.s. normal tissue include VEGFA, pl6INK4A, p53, EGFR, EGF, Ki-67, RAS, NRAS, and cyclin Dl . See, e.g.
Zaravinos et al. "Spotlight on Differentially Expressed Genes in Urinary Bladder Cancer." Cancer Epidemiol Biomarkers Prev. 2009 Feb; 18(2):444-53. Epub 2009 Feb 3. The differential expression of these genes may be indicative of a positive result for cancer.
Nucleic acid biomarkers generally produce a binary result, i.e., presence or absence of an alteration or characteristic in the sample as compared to a healthy control is indicative of a clinical status. Protein biomarkers are generally considered quantitative biomarkers for which a level or amount of the biomarker present in comparison to a reference level or amount indicates a clinical status. As already discussed herein, threshold values for any particular biomarker and associated disease may be determined by reference to literature or standard of care criteria or may be determined empirically.
Amplification refers to production of additional copies of a nucleic acid sequence. See for example, Dieffenbach and Dveksler, PCR Primer, a Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y. (1995), the contents of which is hereby incorporated by reference in its entirety. The amplification reaction may be any amplification reaction known in the art that amplifies nucleic acid molecules, such as polymerase chain reaction, nested polymerase chain reaction, polymerase chain reaction-single strand conformation polymorphism, ligase chain reaction, strand displacement amplification and restriction fragments length polymorphism. In certain methods of the invention, the target nucleic acid and the nucleic acid ligand are PCR amplified. PCR refers to methods by K. B. Mullis (U.S. patent numbers 4,683, 195 and 4,683,202, hereby incorporated by reference) for increasing concentration of a segment of a target sequence in a mixture of genomic DNA without cloning or purification. The process for amplifying the target nucleic acid sequence and nucleic acid ligand includes introducing an excess of oligonucleotide primers that bind the nucleic acid and the nucleic acid ligand, followed by a precise sequence of thermal cycling in the presence of a DNA polymerase. The primers are complementary to their respective strands of the target nucleic acid and nucleic acid ligand.
To effect amplification, the mixture of primers are annealed to their complementary sequences within the target nucleic acid and nucleic acid ligand. Following annealing, the primers are extended with a polymerase so as to form a new pair of complementary strands. The steps of denaturation, primer annealing and polymerase extension can be repeated many times (i.e., denaturation, annealing, and extension constitute one cycle; there can be numerous cycles) to obtain a high concentration of an amplified segment of a desired target and nucleic acid ligand. The length of the amplified segment of the desired target and nucleic acid ligand is determined by relative positions of the primers with respect to each other, and therefore, this length is a controllable parameter.
With PCR, it is possible to amplify a single copy of a specific target sequence in genomic DNA to a level that can be detected by several different methodologies (e.g., staining, hybridization with a labeled probe, incorporation of biotinylated primers followed by avidin- enzyme conjugate detection, incorporation of 32P-labeled deoxynucleotide triphosphates, such as dCTP or dATP, into the amplified segment).
In one embodiment of the invention, the target nucleic acid and nucleic acid ligand can be detected using detectably labeled probes. Nucleic acid probe design and methods of synthesizing oligonucleotide probes are known in the art. See, e.g., Sambrook et al., DNA microarray: A Molecular Cloning Manual, Cold Spring Harbor, N.Y., (2003) or Maniatis, et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, N.Y., (1982), the contents of each of which are herein incorporated by reference herein in their entirety. Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd Ed.), Vols. 1 -3, Cold Spring Harbor Laboratory, (1989) or F. Ausubel et al., Current Protocols In Molecular Biology, Greene Publishing and Wiley- Interscience, New York (1987), the contents of each of which are herein incorporated by reference in their entirety. Suitable methods for synthesizing oligonucleotide probes are also described in Caruthers, Science, 230:281-285, (1985), the contents of which are incorporated by reference.
Probes suitable for use in the present invention include those formed from nucleic acids, such as RNA and/or DNA, nucleic acid analogs, locked nucleic acids, modified nucleic acids, and chimeric probes of a mixed class including a nucleic acid with another organic component such as peptide nucleic acids. Probes can be single stranded or double stranded. Exemplary nucleotide analogs include phosphate esters of deoxyadenosine, deoxycytidine, deoxyguanosine, deoxythymidine, adenosine, cytidine, guanosine, and uridine. Other examples of non-natural nucleotides include a xanthine or hypoxanthine; 5-bromouracil, 2-aminopurine, deoxyinosine, or methylated cytosine, such as 5-methylcytosine, and N4-methoxydeoxycytosine. Also included are bases of polynucleotide mimetics, such as methylated nucleic acids, e.g., 2'-0-methRNA, peptide nucleic acids, modified peptide nucleic acids, and any other structural moiety that can act substantially like a nucleotide or base, for example, by exhibiting base-complementarity with one or more bases that occur in DNA or RNA.
The length of the nucleotide probe is not critical, as long as the probes are capable of hybridizing to the target nucleic acid and nucleic acid ligand. In fact, probes may be of any length. For example, probes may be as few as 5 nucleotides, or as much as 5000 nucleotides. Exemplary probes are 5-mers, 10-mers, 15-mers, 20-mers, 25-mers, 50-mers, 100-mers, 200- mers, 500-mers, 1000-mers, 3000-mers, or 5000-mers. Methods for determining an optimal probe length are known in the art. See, e.g., Shuber, U.S. Patent Number 5,888,778, hereby incorporated by reference in its entirety.
Probes used for detection may include a detectable label, such as a radiolabel, fluorescent label, or enzymatic label. See for example Lancaster et al., U.S. Patent Number 5,869,717, hereby incorporated by reference. In certain embodiments, the probe is fluorescently labeled. Fluorescently labeled nucleotides may be produced by various techniques, such as those described in Kambara et al., Bio/Technol., 6:816-21 , (1988); Smith et al., Nucl. Acid Res., 13:2399-2412, (1985); and Smith et al., Nature, 321 : 674-679, (1986), the contents of each of which are herein incorporated by reference in their entirety. The fluorescent dye may be linked to the deoxyribose by a linker arm that is easily cleaved by chemical or enzymatic means. There are numerous linkers and methods for attaching labels to nucleotides, as shown in Oligonucleotides and Analogues: A Practical Approach, IRL Press, Oxford, (1991); Zuckerman et al., Polynucleotides Res., 15: 5305-5321, ( 1987); Sharma et al., Polynucleotides Res., 19:3019, (1991); Giusti et al., PCR Methods and Applications, 2:223-227, (1993); Fung et al. (U.S. Patent Number 4,757,141); Stabinsky (U.S. Patent Number 4,739,044); Agrawal et al., Tetrahedron Letters, 31 : 1543-1546, (1990); Sproat et al., Polynucleotides Res., 15:4837, (1987); and Nelson et al., Polynucleotides Res., 17:7187-7194, (1989), the contents of each of which are herein incorporated by reference in their entirety. Extensive guidance exists in the literature for derivatizing fluorophore and quencher molecules for covalent attachment via common reactive groups that may be added to a nucleotide. Many linking moieties and methods for attaching fluorophore moieties to nucleotides also exist, as described in Oligonucleotides and Analogues, supra; Guisti et al., supra; Agrawal et al, supra; and Sproat et al., supra
The detectable label attached to the probe may be directly or indirectly detectable. In certain embodiments, the exact label may be selected based, at least in part, on the particular type of detection method used. Exemplary detection methods include radioactive detection, optical absorbance detection, e.g., UV-visible absorbance detection, optical emission detection, e.g., fluorescence; phosphorescence or chemiluminescence; Raman scattering. Preferred labels include optically-detectable labels, such as fluorescent labels. Examples of fluorescent labels include, but are not limited to, 4-acetamido-4'-isothiocyanatostilbene-2,2'disulfonic acid; acridine and derivatives: acridine, acridine isothiocyanate; 5-(2'-aminoethyl)aminonaphthalene-l -sulfonic acid (EDANS); 4-amino-N-[3-vinylsulfonyl)phenyl]naphthalimide-3,5 disulfonate; N-(4-anilino- l -naphthyl)maleimide; anthranilamide; BODIPY; alexa; fluorescien; conjugated multi-dyes; Brilliant Yellow; coumarin and derivatives; coumarin, 7-amino-4-methylcoumarin (AMC, Coumarin 120), 7-amino-4-trifluoromethylcouluarin (Coumaran 151); cyanine dyes; cyanosine; 4',6-diaminidino-2-phenyIindole (DAPI); 5'5"-dibromopyrogallol-sulfonaphthalein
(Bromopyrogallol Red); 7-diethylamino-3-(4'-isothiocyanatophenyl)-4-methylcoumarin;
diethylenetriamine pentaacetate; 4,4'-diisothiocyanatodihydro-stilbene-2,2'-disulfonic acid; 4,4'- diisothiocyanatostilbene-2,2'-disulfonic acid; 5-[dimethylamino]naphthalene-l-sulfonyl chloride (DNS, dansylchloride); 4-dimethylaminophenylazophenyl-4'-isothiocyanate (DABITC); eosin and derivatives; eosin, eosin isothiocyanate, erythrosin and derivatives; erythrosin B, erythrosin, isothiocyanate; ethidium; fluorescein and derivatives; 5-carboxyfluorescein (FAM), 5-(4,6- dichlorotriazin-2-yl)aminofluorescein (DTAF), 2',7'-dimethoxy-4'5'-dichloro-6- carboxyfluorescein, fluorescein, fluorescein isothiocyanate, QFITC, (XRITC); fluorescamine; 1R144; IR1446; Malachite Green isothiocyanate; 4-methylumbelliferoneortho cresolphthalein; nitrotyrosine; pararosaniline; Phenol Red; B-phycoerythrin; o-phthaldialdehyde; pyrene and derivatives: pyrene, pyrene butyrate, succinimidyl 1 -pyrene; butyrate quantum dots; Reactive Red 4 (Cibacron.TM. Brilliant Red 3B-A) rhodamine and derivatives: 6-carboxy-X-rhodamine (ROX), 6-carboxyrhodamine (R6G), lissamine rhodamine B sulfonyl chloride rhodamine (Rhod), rhodamine B, rhodamine 123, rhodamine X isothiocyanate, sulforhodamine B, sulforhodamine 101, sulfonyl chloride derivative of sulforhodamine 101 (Texas Red);
N,N,N',N'tetramethyl-6-carboxyrhodamine (TAMRA); tetramethyl rhodamine; tetramethyl rhodamine isothiocyanate (TRITC); riboflavin; rosolic acid; terbium chelate derivatives; Atto dyes, Cy3; Cy5; Cy5.5; Cy7; IRD 700; IRD 800; La Jolta Blue; phthalo cyanine; and naphthalo cyanine. Labels other than fluorescent labels are contemplated by the invention, including other optically-detectable labels.
Detection of a bound probe may be measured using any of a variety of techniques dependent upon the label used, such as those known to one of skill in the art. Exemplary detection methods include radioactive detection, optical absorbance detection, e.g., UV-visible absorbance detection, optical emission detection, e.g., fluorescence or chemiluminescence. Devices capable of sensing fluorescence from a single molecule include scanning tunneling microscope (siM) and the atomic force microscope (AFM). Hybridization patterns may also be scanned using a CCD camera (e.g., Model TE/CCD512SF, Princeton Instruments, Trenton, N.J.) with suitable optics (Ploem, in Fluorescent and Luminescent Probes for Biological Activity Mason, T. G. Ed., Academic Press, Landon, pp. 1 -1 1 (1993)), such as described in Yershov et al., Proc. Natl. Acad. Sci. 93:4913 (1996), or may be imaged by TV monitoring. For radioactive signals, a phosphorimager device can be used (Johnston et al., Electrophoresis, 13:566, 1990; Drmanac et al., Electrophoresis, 13:566, 1992; 1993). Other commercial suppliers of imaging instruments include General Scanning Inc., (Watertown, Mass. on the World Wide Web at genscan.com), Genix Technologies (Waterloo, Ontario, Canada; on the World Wide Web at confocal.com), and Applied Precision Inc.
In certain embodiments, the target nucleic acid or nucleic acid ligand or both are quantified using methods known in the art. A preferred method for quantitation is quantitative polymerase chain reaction (QPCR). As used herein, "QPCR" refers to a PCR reaction performed in such a way and under such controlled conditions that the results of the assay are quantitative, that is, the assay is capable of quantifying the amount or concentration of a nucleic acid ligand present in the test sample.
QPCR is a technique based on the polymerase chain reaction, and is used to amplify and simultaneously quantify a targeted nucleic acid molecule. QPCR allows for both detection and quantification (as absolute number of copies or relative amount when normalized to DNA input or additional normalizing genes) of a specific sequence in a DNA sample. The procedure follows the general principle of PCR, with the additional feature that the amplified DNA is quantified as it accumulates in the reaction in real time after each amplification cycle. QPCR is described, for example, in Kurnit et al. (U.S. Patent Number 6,033,854), Wang et al. (U.S. Patent Numbers 5,567,583 and 5,348,853), Ma et al. (The Journal of American Science, 2(3), (2006)), Heid et al. (Genome Research 986-994, (1996)), Sambrook and Russell (Quantitative PCR, Cold Spring Harbor Protocols, (2006)), and Higuchi (U.S. Patent Numbers 6,171 ,785 and 5,994,056). The contents of these are incorporated by reference herein in their entirety.
Two common methods of quantification are: (1) use of fluorescent dyes that intercalate with double-stranded DNA, and (2) modified DNA oligonucleotide probes that fluoresce when hybridized with a complementary DNA.
In the first method, a DNA-binding dye binds to all double-stranded (ds)DNA in PCR, resulting in fluorescence of the dye. An increase in DNA product during PCR therefore leads to an increase in fluorescence intensity and is measured at each cycle, thus allowing DNA concentrations to be quantified. The reaction is prepared similarly to a standard PCR reaction, with the addition of fluorescent (ds)DNA dye. The reaction is run in a thermocycler, and after each cycle, the levels of fluorescence are measured with a detector; the dye only fluoresces when bound to the (ds)DNA (i.e., the PCR product). With reference to a standard dilution, the
(ds)DNA concentration in the PCR can be determined. Like other real-time PCR methods, the values obtained do not have absolute units associated with it. A comparison of a measured DNA/RNA sample to a standard dilution gives a fraction or ratio of the sample relative to the standard, allowing relative comparisons between different tissues or experimental conditions. To ensure accuracy in the quantification, it is important to normalize expression of a target gene to a stably expressed gene. This allows for correction of possible differences in nucleic acid quantity or quality across samples. The second method uses sequence-specific RNA or DNA-based probes to quantify only the DNA containing the probe sequence; therefore, use of the reporter probe significantly increases specificity, and allows for quantification even in the presence of some non-specific DNA amplification. This allows for multiplexing, i.e., assaying for several genes in the same reaction by using specific probes with differently colored labels, provided that all genes are amplified with similar efficiency.
This method is commonly carried out with a DNA-based probe with a fluorescent reporter (e.g. 6-carboxyfluorescein) at one end and a quencher (e.g., 6-carboxy- tetramethylrhodamine) of fluorescence at the opposite end of the probe. The close proximity of the reporter to the quencher prevents detection of its fluorescence. Breakdown of the probe by the 5' to 3' exonuclease activity of a polymerase (e.g., Taq polymerase) breaks the reporter- quencher proximity and thus allows unquenched emission of fluorescence, which can be detected. An increase in the product targeted by the reporter probe at each PCR cycle results in a proportional increase in fluorescence due to breakdown of the probe and release of the reporter. The reaction is prepared similarly to a standard PCR reaction, and the reporter probe is added. As the reaction commences, during the annealing stage of the PCR, both probe and primers anneal to the DNA target. Polymerization of a new DNA strand is initiated from the primers, and once the polymerase reaches the probe, its 5 '-3 '-exonuclease degrades the probe, physically separating the fluorescent reporter from the quencher, resulting in an increase in fluorescence. Fluorescence is detected and measured in a real-time PCR thermocycler, and geometric increase of fluorescence corresponding to exponential increase of the product is used to determine the threshold cycle in each reaction.
In certain embodiments, the QPCR reaction uses fluorescent Taqman™ methodology and an instrument capable of measuring fluorescence in real time (e.g., ABI Prism 7700 Sequence Detector; see also PE Biosystems, Foster City, Calif.; see also Gelfand et al., (U.S. Patent Number 5,210,015), the contents of which is hereby incorporated by reference in its entirety). The Taqman™ reaction uses a hybridization probe labeled with two different fluorescent dyes. One dye is a reporter dye (6-carboxyfluorescein), the other is a quenching dye (6-carboxy- tetramethylrhodamine). When the probe is intact, fluorescent energy transfer occurs and the reporter dye fluorescent emission is absorbed by the quenching dye. During the extension phase of the PCR cycle, the fluorescent hybridization probe is cleaved by the 5 '-3' nucleoiytic activity of the DNA polymerase. On cleavage of the probe, the reporter dye emission is no longer transferred efficiently to the quenching dye, resulting in an increase of the reporter dye fluorescent emission spectra.
The nucleic acid ligand of the present invention is quantified by performing QPCR and determining, either directly or indirectly, the amount or concentration of nucleic acid ligand that had bound to its probe in the test sample. The amount or concentration of the bound probe in the test sample is generally directly proportional to the amount or concentration of the nucleic acid ligand quantified by using QPCR. See for example Schneider et al., U.S. Patent Application Publication Number 2009/0042206, Dodge et al., U.S. Patent Number 6,927,024, Gold et al., U.S. Patent Numbers 6,569,620, 6,716,580, and 7,629, 151 , Cheronis et al., U.S. Patent Number 7,074,586, and Ahn et al., U.S. Patent Number 7,642,056, the contents of each of which are herein incorporated by reference in their entirety.
Detecting the presence of the nucleic acid in the analyzed sample directly correlates to the presence of the target protein in that sample. In some embodiments of the invention, the amount of nucleic acid present in the sample correlates to the signal intensity following the conduction of the PCR-based methods. The signal intensity of PCR depends upon the number of PCR cycles performed and/or the starting concentration of the nucleic acid. Since the sequence of the target protein is known to generate the nucleic acid, detection of that specific nucleic acid correlates to the presence of the target protein. Similarly, detection of the amplified target nucleic acid indicates the presence of the target nucleic acid in the sample analyzed.
In one embodiment of the invention, during amplification of the nucleic acid or target nucleic acid using standard PCR methods, one method for detection and quantification of amplified nucleic acid or target nucleic acid results from the presence of a fluorogenic probe. In one embodiment of the invention, the probe, which is specific for the nucleic acid, has a 6- carboxyfluorescein (FAM) moiety covalently bound to the 5-'end and a 6- carboxytetramethylrhodamine (TAMRA) or other fluorescent-quenching dye (easily prepared using standard automated DNA synthesis) present on the 3'-end, along with a 3'-phosphate to prevent elongation. The probe is added with 5'-nuclease to the PCR assays, such that 5'-nuclease cleavage of the probe-nucleic acid duplex results in release of the 5'-bound FAM moiety from the oligonucleotide probe. As amplification continues and more nucleic acid is replicated by the PCR or RT-PCR enzymes, more FAM is released per cycle and so intensity of fluorescence signal per cycle increases. The relative increase in FAM emission is monitored during PCR or RT-PCR amplification using an analytical thermal cycler, or a combined thermal
cycler/laser/detector/software system such as an ABI 7700 Sequence Detector (Applied
Biosystems, Foster City, Calif.). The ABI instrument has the advantage of allowing analysis and display of quantification in less than 60s upon termination of the amplification reactions. Both detection systems employ an internal control or standard wherein a second nucleic acid sequence utilizing the same primers for amplification but having a different sequence and thus different probe, is amplified, monitored and quantitated simultaneously as that for the desired target molecule. See for example, "A Novel Method for Real Time Quantitative RT-PCR," Gibson, U. et. al., 1996, Genome Res. 6:995-1001 ; Piatak, M. et. al., 1993, BioTechniques 14:70-81 ;
"Comparison of the BI 7700 System (TaqMan) and Competitive PCR for Quantification of 1S61 10 DNA in Sputum During Treatment of Tuberculosis," Desjardin, L.e. et. al., 1998, J. Clin. Microbiol. 36(7): 1964-1968), the contents of which are incorporated by reference, herein in their entirety.
In another method for detection and quantification of nucleic acid during amplification, the primers used for amplification contain molecular energy transfer (MET) moieties, specifically fluorescent resonance energy transfer (FRET) moieties, whereby the primers contain both a donor and an acceptor molecule. The FRET pair typically contains a fluorophore donor moiety such as 5-carboxyfluorescein (FAM) or 6-carboxy-4,5-dichloro-2,7- dimethoxyfluorescein (JOE), with an emission maximum of 525 or 546 nm, respectively, paired with an acceptor moiety such as N^TSi'N'-tetramethyl-6-carboxyrhodamine (TAMRA), 6- carboxy-X-rhodamine (ROX) or 6-carboxyrhodamine (R6G), all of which have excitation maximum of 514 nm. The primer may be a hairpin such that the 5'-end of the primer contains the FRET donor, and the 3'-end (based-paired to the 5'-end to form the stem region of the hairpin) contains the FRET acceptor, or quencher. The two moieties in the FRET pair are separated by approximately 15-25 nucleotides in length when the hairpin primer is linearized. While the primer is in the hairpin conformation, no fluorescence is detected. Thus, fluorescence by the donor is only detected when the primer is in a linearized conformation, i.e. when it is incorporated into a double-stranded amplification product. Such a method allows direct quantification of the amount of nucleic acid bound to target molecule in the sample mixture, and this quantity is then used to determine the amount of target molecule originally present in the sample. See for example, Nazarenko, I. A. et al., U.S. Pat. No. 5,866,336, the contents of which is incorporated by reference in its entirety.
In another embodiment of the invention, the QPCR reaction using TaqMan™
methodology selects a TaqMan™ probe based upon the sequence of the nucleic acid to be quantified and generally includes a 5'-end fluor, such as 6-carboxyfluorescein, for example, and a 3'-end quencher, such as, for example, a 6-carboxytetramethylfluorescein, to generate signal as the nucleic acid sequence is amplified using PCR. As the polymerase copies the nucleic acid sequence, the exonuclease activity frees the fluor from the probe, which is annealed downstream from the PCR primers, thereby generating signal. The signal increases as replicative product is produced. The amount of PCR product depends upon both the number of replicative cycles performed as well as the starting concentration of the nucleic acid. In another embodiment, the amount or concentration of an nucleic acid affinity complex (or nucleic acid covalent complex) is determined using an intercalating fluorescent dye during the replicative process. The intercalating dye, such as, for example, SYBR™ green, generates a large fluorescent signal in the presence of double-stranded DNA as compared to the fluorescent signal generated in the presence of single-stranded DNA. As the double-stranded DNA product is formed during PCR, the signal produced by the dye increases. The magnitude of the signal produced is dependent upon both the number of PCR cycles and the starting concentration of the nucleic acid.
Nucleic acids and proteins may be obtained by methods known in the art. Generally, nucleic acids can be extracted from a biological sample by a variety of techniques such as those described by Maniatis, et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, N.Y., pp. 280-281 , (1982), the contents of which is incorporated by reference herein in its entirety. Generally, proteins can be extracted from a biological sample by a variety of techniques such as 2-D electrophoresis, isoelectric focusing, and SDS Slab Gel Electrophoresis. See for example O'Farrell, J. Biol. Chem., 250: 4007-4021 (1975), Sambrook, J. et al., Molecular Cloning: a Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY (1989), Anderson et al., U.S. Patent Number 6,391 ,650, Shepard, U.S. Patent Number 7,229,789, and Han et al., U.S. Patent Number 7,488,579 the contents of each of which is hereby incorporated by reference in its entirety.
In other embodiments, antibodies with a unique oligonucleotide tag are added to the sample to bind a target protein and detection of the oligonucleotide tag results in detection of the protein. The target protein is exposed to an antibody that is coupled to an oligonucleotide tag of a known sequence. The antibody specifically binds the protein, and then PCR is used to amplify the oligonucleotide coupled to the antibody. The identity of the target protein is determined based upon the sequence of the oligonucleotide attached to the antibody and the presence of the oligonucleotide in the sample. In this embodiment of the invention, different antibodies specific for the target protein are used. Each antibody is coupled to a unique oligonucleotide tag of known sequence. Therefore, more than one target protein can be detected in a sample by x identifying the unique oligonucleotide tag attached to the antibody. See for example Kahvejian, U.S. Patent Application Publication Number 2007/0020650, hereby incorporated by reference.
In other embodiments of the invention, antibodies with a unique nucleotide tag are added to the sample to bind the target nucleic acid. As described above, different antibodies specific for the target nucleic acid are used, therefore, more than one target nucleic acid can be detected in a sample by identifying the unique oligonucleotide tag attached. Detection of the nucleotide tag may be done by methods known in the art, such as PCR, QPCR, fluorescent labeling, radiolabeling, biotinylation, Sanger sequencing, sequencing by synthesis, or Single Molecule Real Time Sequencing methods. For description of single molecule sequencing methods see for example, Lapidus, U.S. Patent Number 7,666,593, Quake et al., U.S. Patent Number 7,501,245, and Lapidus et al., U.S. Patent Numbers 7,169,560 and 7,491 ,498, the contents of each of which are herein incorporated by reference.
Antibodies for use in the present invention can be generated by methods well known in the art. See, for example, E. Harlow and D. Lane, Antibodies, a Laboratory Model, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (1988), the contents of which are hereby incorporated by reference in their entirety. In addition, a wide variety of antibodies are available commercially.
The antibody can be obtained from a variety of sources, such as those known to one of skill in the art, including but not limited to polyclonal antibody, monoclonal antibody, monospecific antibody, recombinantly expressed antibody, humanized antibody, plantibodies, and the like; and can be obtained from a variety of animal species, including rabbit, mouse, goat, rat, human, horse, bovine, guinea pig, chicken, sheep, donkey, human, and the like. A wide variety of antibodies are commercially available and a custom-made antibody can be obtained from a number of contract labs. Detailed descriptions of antibodies, including relevant protocols, can be found in, among other places, Current Protocols in Immunology, Coligan et al., eds., John Wiley & Sons (1999, including updates through August 2003); The Electronic Notebook; Basic Methods in Antibody Production and Characterization, G. Howard and D. Bethel, eds., CRC Press (2000); J. Coding, Monoclonal Antibodies: Principles and Practice, 3d Ed., Academic Press (1996); E. Harlow and D. Lane, Using Antibodies, Cold Spring Harbor Lab Press (1999); P. Shepherd and C. Dean, Monoclonal Antibodies: A Practical Approach, Oxford University Press (2000); A. Johnstone and M. Turner, Immunochemistry 1 and 2, Oxford University Press (1997); C. Borrebaeck, Antibody Engineering, 2d ed., Oxford university Press (1995); A.
Johnstone and R. Thorpe, Immunochemistry in Practice, Blackwell Science, Ltd. (1996); H. Zola, Monoclonal Antibodies: Preparation and Use of Monoclonal Antibodies and Engineered Antibody Derivatives (Basics: From Background to Bench), Springer Verlag (2000); and S. Hockfield et al., Selected Methods for Antibody and Nucleic Acid Probes, Cold Spring Harbor Lab Press (1993).
In certain embodiments, the target nucleic acid or nucleic acid ligand or both are detected using sequencing. In those embodiments, the nucleic acid/protein complex may be dissociated, releasing the nucleic acid for the sequencing reaction. Sequencing-by-synthesis is a common technique used in next generation procedures and works well with the instant invention.
However, other sequencing methods can be used, including sequence-by-ligation, sequencing- by-hybridization, gel-based techniques and others. In general, sequencing involves hybridizing a primer to a template to form a template/primer duplex, contacting the duplex with a polymerase in the presence of a detectably-labeled nucleotides under conditions that permit the polymerase to add nucleotides to the primer in a template-dependent manner. Signal from the detectable label is then used to identify the incorporated base and the steps are sequentially repeated in order to determine the linear order of nucleotides in the template. Exemplary detectable labels include radiolabels, florescent labels, enzymatic labels, etc. In particular embodiments, the detectable label may be an optically detectable label, such as a fluorescent label. Exemplary fluorescent labels include cyanine, rhodamine, fluorescien, coumarin, BODIPY, alexa, or conjugated multi-dyes. Numerous techniques are known for detecting sequences and some are exemplified below. However, the exact means for detecting and compiling sequence data does not affect the function of the invention described herein. In a preferred embodiment, the target nucleic acids, nucleic acid ligands, or both are detected using single molecule sequencing. Advantageously, methods of the invention have found that single molecule sequencing of DNA or protein biomarkers (via nucleic acid ligands) from urine samples show an increased sensitivity as compared to qPCR-based assays of biomarkers from urine samples. In fact, single molecule sequencing of DNA and protein biomarkers in urine has comparable sensitivity as qPCR sequencing of DNA and protein biomarkers from tissue samples, as highlighted in Example 3 below. Accordingly, assays of the invention that detect biomarkers in urine samples have similar performance and sensitivity of invasive tissue-based assays.
An example of a single molecule sequencing technique suitable for use in the methods of the provided invention is Ion Torrent sequencing (U.S. patent application numbers
2009/0026082, 2009/0127589, 2010/0035252, 2010/0137143, 2010/0188073, 2010/0197507, 2010/0282617, 2010/0300559), 2010/0300895, 2010/0301398, and 2010/0304982), the content of each of which is incorporated by reference herein in its entirety. In Ion Torrent sequencing, DNA is sheared into fragments of approximately 300-800 base pairs, and the fragments are blunt ended. Oligonucleotide adaptors are then ligated to the ends of the fragments. The adaptors serve as primers for amplification and sequencing of the fragments. The fragments can be attached to a surface and is attached at a resolution such that the fragments are individually resolvable. Addition of one or more nucleotides releases a proton (H+), which signal detected and recorded in a sequencing instrument. The signal strength is proportional to the number of nucleotides incorporated. User guides describe in detail the Ion Torrent protocol(s) that are suitable for use in methods of the invention, such as Life Technologies' literature entitled "Ion Sequencing Kit for User Guide v. 2.0" for use with their sequencing platform the Personal Genome MachineTM (PCG).
In one embodiment, single molecule sequencing is used to maximize detection of FGFR3 mutations by conducting the biomarker assay on the Ion Torrent PGM platform (Life
Technologies) ultra-deep sequencing platform. A primary PCR step is carried out using chimeric primers containing a sequence specific portion for amplifying the exons of interest (Exons 7, 10, and 15) along with adapter sequences required for sequencing analysis. Sequence specific primers suitable for use in smFGFR3 can be designed using any method known in the art. In certain embodiments, the primer can vary in lengths between 16bp to 22 bp. The primary consideration is the Tm of the sequence specific portion. For example, primers with target specific Tm values ranging from ~52°C to ~68°C generated successful amplification products with chimeric oligonucleotides. Another consideration for primer design is the size of the amplicon because PCR products generated from total urine DNA have decreased yields at sizes larger than 300bp. Accordingly, in certain embodiments, FGFR3 amplicons are designed to be ~100bp or smaller to accommodate read lengths on the sequencing platform. Although the above example is directed towards single molecule detection of FGFR3, methods of the invention also provide for single molecule detection of other nucleic acids, such as TWIST1 , VIM, and NID2, and proteins such as MMP-2, MMP-9, and ADAM- 12, through detection of protein-specific nucleic acids.
Another example of a DNA sequencing technique that can be used in the methods of the provided invention is 454 sequencing (Roche) (Margulies, M et al. 2005, Nature, 437, 376-380). 454 sequencing involves two steps. In the first step, DNA is sheared into fragments of approximately 300-800 base pairs, and the fragments are blunt ended. Oligonucleotide adaptors are then ligated to the ends of the fragments. The adaptors serve as primers for amplification and sequencing of the fragments. The fragments can be attached to DNA capture beads, e.g., streptavidin-coated beads using, e.g., Adaptor B, which contains 5'-biotin tag. The fragments attached to the beads are PCR amplified within droplets of an oil-water emulsion. The result is multiple copies of clonally amplified DNA fragments on each bead. In the second step, the beads are captured in wells (pico-liter sized). Pyrosequencing is performed on each DNA fragment in parallel. Addition of one or more nucleotides generates a light signal that is recorded by a CCD camera in a sequencing instrument. The signal strength is proportional to the number of nucleotides incorporated. Pyrosequencing makes use of pyrophosphate (PPi) which is released upon nucleotide addition. PPi is converted to ATP by ATP sulfurylase in the presence of adenosine 5' phosphosulfate. Luciferase uses ATP to convert luciferin to oxyluciferin, and this reaction generates light that is detected and analyzed.
Another example of a DNA sequencing technique that can be used in the methods of the provided invention is SOLiD technology (Applied Biosystems). In SOLiD sequencing, genomic DNA is sheared into fragments, and adaptors are attached to the 5' and 3' ends of the fragments to generate a fragment library. Alternatively, internal adaptors can be introduced by ligating adaptors to the 5' and 3' ends of the fragments, circularizing the fragments, digesting the circularized fragment to generate an internal adaptor, and attaching adaptors to the 5' and 3' ends of the resulting fragments to generate a mate-paired library. Next, clonal bead populations are prepared in microreactors containing beads, primers, template, and PCR components. Following PCR, the templates are denatured and beads are enriched to separate the beads with extended templates. Templates on the selected beads are subjected to a 3' modification that permits bonding to a glass slide. The sequence can be determined by sequential hybridization and ligation of partially random oligonucleotides with a central determined base (or pair of bases) that is identified by a specific fluorophore. After a color is recorded, the ligated oligonucleotide is cleaved and removed and the process is then repeated.
Another example of a sequencing technology that can be used in the methods of the provided invention is Illumina sequencing. Illumina sequencing is based on the amplification of DNA on a solid surface using fold-back PCR and anchored primers. Genomic DNA is fragmented, and adapters are added to the 5' and 3' ends of the fragments. DNA fragments that are attached to the surface of flow cell channels are extended and bridge amplified. The fragments become double stranded, and the double stranded molecules are denatured. Multiple cycles of the solid-phase amplification followed by denaturation can create several million clusters of approximately 1 ,000 copies of single-stranded DNA molecules of the same template in each channel of the flow cell. Primers, DNA polymerase and four fluorophore-labeled, reversibly terminating nucleotides are used to perform sequential sequencing. After nucleotide incorporation, a laser is used to excite the fluorophores, and an image is captured and the identity of the first base is recorded. The 3' terminators and fluorophores from each incorporated base are removed and the incorporation, detection and identification steps are repeated.
Another example of a sequencing technology that can be used in the methods of the provided invention includes the single molecule, real-time (SMRT) technology of Pacific Biosciences. In SMRT, each of the four DNA bases is attached to one of four different fluorescent dyes. These dyes are phospholinked. A single DNA polymerase is immobilized with a single molecule of template single stranded DNA at the bottom of a zero-mode waveguide (ZMW). A ZMW is a confinement structure which enables observation of incorporation of a single nucleotide by DNA polymerase against the background of fluorescent nucleotides that rapidly diffuse in an out of the ZMW (in microseconds). It takes several milliseconds to incorporate a nucleotide into a growing strand. During this time, the fluorescent label is excited and produces a fluorescent signal, and the fluorescent tag is cleaved off. Detection of the corresponding fluorescence of the dye indicates which base was incorporated. The process is repeated.
Another example of a sequencing technique that can be used in the methods of the provided invention is nanopore sequencing (Soni G V and Meller A. (2007) Clin Chem 53: 996-2001). A nanopore is a small hole, of the order of 1 nanometer in diameter. Immersion of a nanopore in a conducting fluid and applicatiorrof a potential across it results in a slight electrical current due to conduction of ions through the nanopore. The amount of current which flows is sensitive to the size of the nanopore. As a DNA molecule passes through a nanopore, each nucleotide on the DNA molecule obstructs the nanopore to a different degree. Thus, the change in the current passing through the nanopore as the DNA molecule passes through the nanopore represents a reading of the DNA sequence.
Another example of a sequencing technique that can be used in the methods of the provided invention involves using a chemical-sensitive field effect transistor (chemFET) array to sequence DNA (for example, as described in US Patent Application Publication No.
20090026082). In one example of the technique, DNA molecules can be placed into reaction chambers, and the template molecules can be hybridized to a sequencing primer bound to a polymerase. Incorporation of one or more triphosphates into a new nucleic acid strand at the 3' end of the sequencing primer can be detected by a change in current by a chemFET. An array can have multiple chemFET sensors. In another example, single nucleic acids can be attached to beads, and the nucleic acids can be amplified on the bead, and the individual beads can be transferred to individual reaction chambers on a chemFET array, with each chamber having a chemFET sensor, and the nucleic acids can be sequenced.
Another example of a sequencing technique that can be used in the methods of the provided invention involves using an electron microscope (Moudrianakis E. N. and Beer M. Proc Natl Acad Sci USA. 1965 March; 53:564-71). In one example of the technique, individual DNA molecules are labeled using metallic labels that are distinguishable using an electron microscope. These molecules are then stretched on a flat surface and imaged using an electron microscope to measure sequences.
In certain embodiments, methods of the invention provide for detection of methylation patterns in nucleic acids. Methods include a number of bisulfite treatment sequencing methods in which genomic DNA is isolated and treated with bisulfite. Bisulfite DNA sequencing utilizes bisulfite-induced modification of genomic DNA under conditions whereby unmethylated cytosine is converted to uracil. The bisulfite-modified sequence is then amplified by PCR with two sets of strand-specific primers to yield a pair of fragments, one from each strand, in which all uracil and thymine residues are amplified as thymine and only 5-methylcytosine residues are amplified as cytosine; The PCR products can be sequenced or can be cloned and sequenced to provide methylation maps of single DNA molecules. See Frommer, M. et al., Proc. Natl. Acad. Sci. 89: 1827-1831 (1992). In certain aspects, after the nucleic acids are bisulfite modified, a barcode be ligated to the bisulfite modified targets and the methylated sample library can be pooled with other target nucleic acids and/or nucleic acids for multiplex sequencing.
Perhaps the most widely-used method of probing methylation patterns is methylation specific PCR (MSP) which uses two sets of primers for an amplification reaction. One primer set is complimentary to sequences whose Cs are converted to Us by bisulfite, and the other primer set is complimentary to non-converted Cs. Using these two separate primer sets, both the methylated and unmethylated DNA are amplified. Comparison of the amplification products gives insight as to the methylation in a given sequence. See Herman et al., "Methylation-specific PCR: A novel PCR assay for methylation status of CpG islands," P.N.A.S., vol. 93, p. 9821-26 (1996), which is incorporated herein by reference in its entirety. This technique can detect methylation changes as small as ± 0.1%. In addition to methylation of CpG islands, many of the sequences surrounding clinically relevant hypermethylated CpG islands can also be
hypermethylated, and are potential biomarkers.
Beyond MSP, it is also possible to measure methylation levels by using hybridization probes that are specific for the products of bisulfate-converted nucleic acids using real-time PCR with primers that not complimentary to the CpG island regions of interest, or primers that hybridize to sequences adjacent to the CpG islands. Methods of using primers having abasic and or mismatch regions corresponding to CpG islands are disclosed in U.S. Patent Application No. 13/472,209 "Primers for Analyzing Methylated Sequences and Methods of Use Thereof," filed May 15, 2012, and incorporated by reference herein in its entirety. Additionally, it is possible to determine an amount of methylation by amplifying and directly sequencing nucleic acids by using single molecule sequencing. Sequences can be read that originate from a single molecule or that originate from amplifications from a single molecule. Millions of independent amplifications of single molecules can be performed in parallel either on a solid surface or in tiny compartments in water/oil emulsion. The DNA sample to be sequenced can be diluted and/or dispersed sufficiently to obtain one molecule in each compartment. This dilution can be followed by DNA amplification to generate copies of the original DNA sequences and creating "clusters" of molecules all having the same sequence. These clusters can then be sequenced. Many millions of reads can be generated in one run. Sequence can be generated starting at the 5' end of a given strand of an amplified sequence and/or sequence can be generated from starting from the 5' end of the complementary sequence. In a preferred embodiment, sequence from strands is generated, i.e. paired end reads (see for example, Harris, U.S. patent number 7,767,400).
Nucleotides useful in the invention include any nucleotide or nucleotide analog, whether naturally-occurring or synthetic. For example, preferred nucleotides include phosphate esters of deoxyadenosine, deoxycytidine, deoxyguanosine, deoxythymidine, adenosine, cytidine, guanosine, and uridine. Other nucleotides useful in the invention comprise an adenine, cytosine, guanine, thymine base, a xanthine or hypoxanthine; 5-bromouracil, 2-aminopurine,
deoxyinosine, or methylated cytosine, such as 5-methylcytosine, and N4-methoxydeoxycytosine. Also included are bases of polynucleotide mimetics, such as methylated nucleic acids, e.g., 2'-0- methRNA, peptide nucleic acids, modified peptide nucleic acids, locked nucleic acids and any other structural moiety that can act substantially like a nucleotide or base, for example, by exhibiting base-complementarity with one or more bases that occur in DNA or RNA and/or being capable of base-complementary incorporation, and includes chain-terminating analogs. A nucleotide corresponds to a specific nucleotide species if they share base-complementarity with respect to at least one base.
Nucleotides for nucleic acid sequencing according to the invention preferably include a detectable label that is directly or indirectly detectable. Preferred labels include optically- detectable labels, such as fluorescent labels. Examples of fluorescent labels include, but are not limited to, 4-acetamido-4'-isothiocyanatostilbene-2,2'disulfonic acid; acridine and derivatives: acridine, acridine isothiocyanate; 5-(2'-aminoethyl)aminonaphthalene-l-sulfonic acid (EDANS); 4-amino-N-[3-vinylsulfonyl)phenyl]naphthalimide-3,5 disulfonate; N-(4-anilino-l- naphthyl)maleimide; anthranilamide; BODIPY; Brilliant Yellow; coumarin and derivatives; coumarin, 7-amino-4-methylcoumarin (AMC, Coumarin 120), 7-amino-4- trifluoromethylcouluarin (Coumaran 151); cyanine dyes; cyanosine; 4',6-diaminidino-2- phenylindole (DAPI); 5'5"-dibromopyrogallol-sulfonaphthalein (Bromopyrogallol Red); 7- diethylamino-3-(4'-isothiocyanatophenyl)-4-methylcoumarin; diethylenetriamine pentaacetate; 4,4'-diisothiocyanatodihydro-stilbene-2,2'-disulfonic acid; 4,4'-diisothiocyanatostilbene-2,2'- disulfonic acid; 5-[dimethylamino]naphthalene-l-sulfonyl chloride (DNS, dansylchloride); 4- dimethylaminophenylazophenyl-4'-isothiocyanate (DABITC); eosin and derivatives; eosin, eosin isothiocyanate, erythrosin and derivatives; erythrosin B, erythrosin, isothiocyanate; ethidium; fluorescein and derivatives; 5-carboxyfluorescein (FAM), 5-(4,6-dichlorotriazin-2- yl)aminofluorescein (DTAF), 2',7'-dimethoxy-4'5'-dichloro-6-carboxyfluorescein, fluorescein, fluorescein isothiocyanate, QFITC, (XRITC); fluorescamine; IR144; IR1446; Malachite Green isothiocyanate; 4-methylumbelliferoneortho cresolphthalein; nitrotyrosine; pararosaniline; Phenol Red; B-phycoerythrin; o-phthaldialdehyde; pyrene and derivatives: pyrene, pyrene butyrate, succinimidyl 1 -pyrene; butyrate quantum dots; Reactive Red 4 (Cibacron.TM. Brilliant Red 3B-A) rhodamine and derivatives: 6-carboxy-X-rhodamine (ROX), 6-carboxyrhodamine (R6G), lissamine rhodamine B sulfonyl chloride rhodamine (Rhod), rhodamine B, rhodamine 123, rhodamine X isothiocyanate, sulforhodamine B, sulforhodamine 101 , sulfonyl chloride derivative of sulforhodamine 101 (Texas Red); N,N,N',N'tetramethyl-6-carboxyrhodamine (TAMRA); tetramethyl rhodamine; tetramethyl rhodamine isothiocyanate (TRITC); riboflavin; rosolic acid; terbium chelate derivatives; Cy3; Cy5; Cy5.5; Cy7; IRD 700; IRD 800; La Jolta Blue; phthalo cyanine; and naphthalo cyanine. Preferred fluorescent labels are cyanine-3 and cyanine-5. Labels other than fluorescent labels are contemplated by the invention, including other optically-detectable labels.
Nucleic acid polymerases generally useful in the invention include DNA polymerases, RNA polymerases, reverse transcriptases, and mutant or altered forms of any of the foregoing. DNA polymerases and their properties are described in detail in, among other places, DNA Replication 2nd edition, Kornberg and Baker, W. H. Freeman, New York, N.Y. (1991). Known conventional DNA polymerases useful in the invention include, but are not limited to,
Pyrococcus furiosus (Pfu) DNA polymerase (Lundberg et al., 1991 , Gene, 108: 1 , Stratagene), Pyrococcus woesei (Pwo) DNA polymerase (Hinnisdaels et al., 1996, Biotechniques, 20: 186-8, Boehringer Mannheim), Thermus thermophilus (Tth) DNA polymerase (Myers and Gelfand 1991 , Biochemistry 30:7661), Bacillus stearothermophilus DNA polymerase (Stenesh and McGowan, 1977, Biochim Biophys Acta 475:32), Thermococcus litoralis (Tli) DNA polymerase (also referred to as Vent.TM. DNA polymerase, Cariello et al., 1991, Polynucleotides Res, 19: 4193, New England Biolabs), 9.degree.Nm.TM. DNA polymerase (New England Biolabs), Stoffel fragment, ThermoSequenase® (Amersham Pharmacia Biotech UK), Therminator.TM. (New England Biolabs), Thermotoga maritima (Tma) DNA polymerase (Diaz and Sabino, 1998 Braz J; Med. Res, 31 : 1239), Thermus aquaticus (Taq) DNA polymerase (Chien et al., 1976, J. Bacteoriol, 127: 1550), DNA polymerase, Pyrococcus kodakaraensis KOD DNA polymerase (Takagi et al., 1997, Appl. Environ. Microbiol. 63:4504), JDF-3 DNA polymerase (from thermococcus sp. JDF-3, Patent application WO 0132887), Pyrococcus GB-D (PGB-D) DNA polymerase (also referred as Deep Vent.TM. DNA polymerase, Juncosa-Ginesta et al., 1994, Biotechniques, 16:820, New England Biolabs), UlTma DNA polymerase (from thermophile Thermotoga maritima; Diaz and Sabino, 1998 Braz J. Med. Res, 31 : 1239; PE Applied
Biosystems), Tgo DNA polymerase (from thermococcus gorgonarius, Roche Molecular
Biochemicals), E. coli DNA polymerase I (Lecomte and Doubleday, 1983, Polynucleotides Res. 1 1 :7505), T7 DNA polymerase (Nordstrom et al., 1981 , J. Biol. Chem. 256:31 12), and archaeal DP1 I/DP2 DNA polymerase II (Cann et al, 1998, Proc. Natl. Acad. Sci. USA 95: 14250).
Both mesophilic polymerases and thermophilic polymerases are contemplated.
Thermophilic DNA polymerases include, but are not limited to, ThermoSequenase®,
9.degree.Nm.TM., Therminator.TM., Taq, Tne, Tma, Pfu, Tfl, Tth, Tli, Stoffel fragment, Vent.TM. and Deep Vent.TM. DNA polymerase, KOD DNA polymerase, Tgo, JDF-3, and mutants, variants and derivatives thereof. A highly-preferred form of any polymerase is a 3' exonuclease-deficient mutant.
Reverse transcriptases useful in the invention include, but are not limited to, reverse transcriptases from HIV, HTLV-1, HTLV-II, FeLV, FIV, SIV, AMV, MMTV, MoMuLV and other retroviruses (see Levin, Cell 88:5-8 (1997); Verma, Biochim Biophys Acta. 473:1-38 (1977); Wu et al., CRC Crit. Rev Biochem. 3:289-347 (1975)).
In a preferred embodiment, nucleic acid template molecules are attached to a substrate (also referred to herein as a surface) and subjected to analysis by single molecule sequencing as described herein. Nucleic acid template molecules are attached to the surface such that the template/primer duplexes are individually optically resolvable. Substrates for use in the invention can be two- or three-dimensional and can comprise a planar surface (e.g., a glass slide) or can be shaped. A substrate can include glass (e.g., controlled pore glass (CPG)), quartz, plastic (such as polystyrene (low cross-linked and high cross-linked polystyrene), polycarbonate, polypropylene and poly(methymethacrylate)), acrylic copolymer, polyamide, silicon, metal (e.g., alkanethiolate- derivatized gold), cellulose, nylon, latex, dextran, gel matrix (e.g., silica gel), polyacrolein, or composites.
Suitable three-dimensional substrates include, for example, spheres, microparticles, beads, membranes, slides, plates, micromachined chips, tubes (e.g., capillary tubes), microwells, microfluidic devices, channels, filters, or any other structure suitable for anchoring a nucleic acid. Substrates can include planar arrays or matrices capable of having regions that include populations of template nucleic acids or primers. Examples include nucleoside-derivatized CPG and polystyrene slides; derivatized magnetic slides; polystyrene grafted with polyethylene glycol, and the like.
Substrates are preferably coated to allow optimum optical processing and nucleic acid attachment. Substrates for use in the invention can also be treated to reduce background.
Exemplary coatings include epoxides, and derivatized epoxides (e.g., with a binding molecule, such as an oligonucleotide or streptavidin).
Various methods can be used to anchor or immobilize the nucleic acid molecule to the surface of the substrate. The immobilization can be achieved through direct or indirect bonding to the surface. The bonding can be by covalent linkage. See, Joos et al., Analytical Biochemistry 247:96-101 , 1997; Oroskar et al., Clin. Chem. 42: 1547-1555, 1996; and handjian, Mol. Bio. Rep. 1 1 : 107-1 15, 1986. A preferred attachment is direct amine bonding of a terminal nucleotide of the template or the 5' end of the primer to an epoxide integrated on the surface. The bonding also can be through non-covalent linkage. For example, biotin-streptavidin (Taylor et al., J. Phys. D. Appl. Phys. 24: 1443, 1991) and digoxigenin with anti-digoxigenin (Smith et al., Science 253: 1 122, 1992) are common tools for anchoring nucleic acids to surfaces and parallels.
Alternatively, the attachment can be achieved by anchoring a hydrophobic chain into a lipid monolayer or bilayer. Other methods for known in the art for attaching nucleic acid molecules to substrates also can be used.
Any detection method can be used that is suitable for the type of label employed. Thus, exemplary detection methods include radioactive detection, optical absorbance detection, e.g., UV-visible absorbance detection, optical emission detection, e.g., fluorescence or chemiluminescence. For example, extended primers can be detected on a substrate by scanning all or portions of each substrate simultaneously or serially, depending on the scanning method used. For fluorescence labeling, selected regions on a substrate may be serially scanned one-by- one or row-by-row using a fluorescence microscope apparatus, such as described in Fodor (U.S. Pat. No. 5,445,934) and Mathies et al. (U.S. Pat. No. 5,091 ,652). Devices capable of sensing fluorescence from a single molecule include scanning tunneling microscope (siM) and the atomic force microscope (AFM). Hybridization patterns may also be scanned using a CCD camera (e.g., Model TE/CCD512SF, Princeton Instruments, Trenton, N.J.) with suitable optics (Ploem, in Fluorescent and Luminescent Probes for Biological Activity Mason, T. G. Ed., Academic Press, Landon, pp. 1 -1 1 (1993), such as described in Yershov et al., Proc. Natl. Acad. Sci. 93:4913 (1996), or may be imaged by TV monitoring. For radioactive signals, a phosphorimager device can be used (Johnston et al., Electrophoresis, 13:566, 1990; Drmanac et al., Electrophoresis, 13:566, 1992; 1993). Other commercial suppliers of imaging instruments include General Scanning Inc., (Watertown, Mass. on the World Wide Web at genscan.com), Genix
Technologies (Waterloo, Ontario, Canada; on the World Wide Web at confocal.com), and Applied Precision Inc. Such detection methods are particularly useful to achieve simultaneous scanning of multiple attached template nucleic acids.
A number of approaches can be used to detect incorporation of fluorescently-labeled nucleotides into a single nucleic acid molecule. Optical setups include near-field scanning microscopy, far-field confocal microscopy, wide-field epi-illumination, light scattering, dark field microscopy, photoconversion, single and/or multiphoton excitation, spectral wavelength discrimination, fluorophor identification, evanescent wave illumination, and total internal reflection fluorescence (TIRF) microscopy. In general, certain methods involve detection of laser-activated fluorescence using a microscope equipped with a camera. Suitable photon detection systems include, but are not limited to, photodiodes and intensified CCD cameras. For example, an intensified charge couple device (ICCD) camera can be used. The use of an ICCD camera to image individual fluorescent dye molecules in a fluid near a surface provides numerous advantages. For example, with an ICCD optical setup, it is possible to acquire a sequence of images (movies) of fluorophores. Some embodiments of the present invention use TIRF microscopy for imaging. TIRF microscopy uses totally internally reflected excitation light and is well known in the art. See, e.g., the World Wide Web at nikon-instruments.jp/eng/page/products/tirf.aspx. In certain embodiments, detection is carried out using evanescent wave illumination and total internal reflection fluorescence microscopy. An evanescent light field can be set up at the surface, for example, to image fluorescently-labeled nucleic acid molecules. When a laser beam is totally reflected at the interface between a liquid and a solid substrate (e.g., a glass), the excitation light beam penetrates only a short distance into the liquid. The optical field does not end abruptly at the reflective interface, but its intensity falls off exponentially with distance. This surface electromagnetic field, called the "evanescent wave", can selectively excite fluorescent molecules in the liquid near the interface. The thin evanescent optical field at the interface provides low background and facilitates the detection of single molecules with high signal-to-noise ratio at visible wavelengths.
The evanescent field also can image fluorescently-labeled nucleotides upon their incorporation into the attached template/primer complex in the presence of a polymerase. Total internal reflectance fluorescence microscopy is then used to visualize the attached
template/primer duplex and/or the incorporated nucleotides with single molecule resolution.
Some embodiments of the invention use non-optical detection methods such as, for example, detection using nanopores (e.g., protein or solid state) through which molecules are individually passed so as to allow identification of the molecules by noting characteristics or changes in various properties or effects such as capacitance or blockage current flow (see, for example, Stoddart et al, Proc. Nat. Acad. Sci., 106:7702, 2009; Purnell and Schmidt, ACS Nano, 3 :2533, 2009; Branton et al, Nature Biotechnology, 26: 1 146, 2008; Polonsky et al, U.S.
Application 2008/0187915; Mitchell & Howorka, Angew. Chem. Int. Ed. 47:5565, 2008;
Borsenberger et al, J. Am. Chem. Soc, 131 , 7530, 2009) ; or other suitable non-optical detection methods.
Alignment and/or compilation of sequence results obtained from the image stacks produced as generally described above utilizes look-up tables that take into account possible sequences changes (due, e.g., to errors, mutations, etc.). Essentially, sequencing results obtained as described herein are compared to a look-up type table that contains all possible reference sequences plus 1 or 2 base errors. In some embodiments, a plurality of nucleic acid molecules being sequenced is bound to a solid support. To immobilize the nucleic acid on a solid support, a capture sequence/universal priming site can be added at the 3' and/or 5' end of the template. The nucleic acids may be bound to the solid support by hybridizing the capture sequence to a complementary sequence covalently attached to the solid support. The capture sequence (also referred to as a universal capture sequence) is a nucleic acid sequence complimentary to a sequence attached to a solid support that may dually serve as a universal primer. In some embodiments, the capture sequence is polyNn, wherein N is U, A, T, G, or C, e g., 20-70, 40-60, e.g., about 50. For example, the capture sequence could be polyT40-50 or its complement. As an alternative to a capture sequence, a member of a coupling pair (such as, e.g., antibody/antigen, receptor/ligand, or the avidin-biotin pair as described in, e.g., U.S. Patent Application No. 2006/0252077) may be linked to each fragment to be captured on a surface coated with a respective second member of that coupling pair.
In some embodiments, a barcode sequence is attached to the nucleic acid, the nucleic acid, or both. See for example, Steinman et al. (PCT internal application number
PCT/US09/64001), the content of which is incorporated by reference herein in its entirety.
Kits
In one embodiment the present invention relates to a kit comprising a detection reagent which binds to any nucleic acid sequence of ADAM 12, GSTPl , FGFR3, MMP2, TWISTI, NID2, Vimentin, and/or p53, and/or polypeptides encoded thereby for the determination of bladder cancer.
One embodiment of the present invention relates to a kit for screening for, assessing the prognosis of an individual with bladder cancer, which comprises a reagent selected from the group consisting of: (a) a reagent for detecting mRNA of the ADAM12, GSTPl, FGFR3, MMP2, TWISTI , NID2, Vimentin, and/or p53 gene; (b) a reagent for detecting protein levels of ADAM 12, GSTPl, FGFR3, MMP2, TWISTI, NID2, Vimentin, and/or p53; and (c) a reagent for detecting the biological activity of the ADAM 12, GSTPl , FGFR3, MMP2, TWISTI, NID2, Vimentin, and/or p53.
In one embodiment, the present invention provides kits for detecting one or more of the following: a mutation in the FGFR3 gene, methylation status of TWISTI, methylation status of NID2, methylation status of Vimentin, protein levels of MMP2, a loss of heterozygozity in p53, and expression levels of ADAM12 protein. Further embodiments of kits may include additional biomarkers. In certain embodiments, the present invention provides kits for measuring the expression of the protein and/or RNA products of ADAM 12, GSTPl , FGFR3, MMP2, TWIST 1 , NID2, Vimentin, and/or p53 in combination with at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, all or any combinational biomarkers mentioned herein.
Kits encompassed by the invention comprise materials and reagents required for measuring the expression of such protein and RNA products. In specific embodiments, the kits may further comprise one or more additional reagents employed in the various methods, such as: (1) reagents for stabilizing and/or purifying RNA from the sample (2) primers for generating test nucleic acids; (3) dNTPs and/or rNTPs (either premixed or separate), optionally with one or more uniquely labelled dNTPs and/or rNTPs (e.g., biotinylated or Cy3 or Cy5 tagged dNTPs);
(4) post synthesis labelling reagents, such as chemically active derivatives of fluorescent dyes;
(5) enzymes, such as reverse transcriptases, DNA polymerases, and the like; (6) various buffer mediums, e.g., reaction, hybridization and washing buffers; (7) labelled probe purification reagents and components, like spin columns, etc.; and (8) protein purification reagents; (9) signal generation and detection reagents, e.g., streptavidin-alkaline phosphatase conjugate,
chemifluorescent or chemiluminescent substrate, and the like.
In particular embodiments, the kits comprise prelabeled quality controlled protein and or RNA isolated from a sample (e.g., blood or chondrocytes or synovial fluid) for use as a control. In some embodiments, the kits are RT-PCR or qRT-PCR kits. In other embodiments, the kits are nucleic acid arrays and protein arrays. Such kits according to the subject invention will at least comprise an array having associated protein or nucleic acid members of the invention and packaging means therefore. Alternatively, the protein or nucleic acid members of the invention may be pre-packaged onto an array.
In some embodiments, the kits are quantitative RT-PCR kits. In one embodiment, the quantitative RT-PCR kit includes the following: (a) primers used to amplify each of a combination of biomarkers of the invention; (b) buffers and enzymes including an reverse transcriptase; (c) one or more thermos table polymerases; and (d) Sybr® Green. In another embodiment, the kit of the invention also includes (a) a reference control RNA and (b) a spiked control RNA.
The invention provides kits that are useful for (a) diagnosing individuals as having bladder cancer and/or early stage bladder cancer. The invention also provides kits that are useful for determining the likelihood of bladder cancer in patients presented with hematuria. Additional embodiments of the invention include kits that are useful for monitoring the recurrence of bladder cancer. For example, in a particular embodiment of the invention a kit is comprised a forward and reverse primer wherein the forward and reverse primer are designed to quantitate expression of all of the species of mRNA corresponding to each of the biomarkers as identified in accordance with the invention useful in determining whether an individual has bladder cancer and/or early stage bladder cancer or not. In certain embodiments, at least one of the primers is designed to span an exon junction.
The invention provides kits that are useful for detecting, diagnosing, monitoring and prognosing bladder cancer based upon the detection of protein or RNA products of AD AMI 2, GSTP1 , FGFR3, MMP2, TWIST1, NID2, Vimentin, and/or p53, possibly in combination with at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, all or any combination of the combinatorial biomarkers of the invention in a sample.
In certain embodiments, such kits do not include the materials and reagents for measuring the expression of a protein or RNA product of a biomarker of the invention that has been suggested by the prior art to be associated with bladder cancer. In other embodiments, such kits include the materials and reagents for measuring the expression of a protein or RNA product of a combinatorial biomarker of the invention that has been suggested by the prior art to be associated with bladder cancer and at least 1 , at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45 or more genes other than the combinatorial biomarkers of the invention.
The invention provides kits useful for monitoring the efficacy of one or more therapies that a subject is undergoing based upon detecting a protein or RNA product of ADAM 12, GSTP1 , FGFR3, MMP2, TWIST1, NID2, Vimentin, and/or p53, possibly in combination with any number of up to at least 1 , at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, all or any combination of the combinatorial biomarkers of the invention in a sample. In certain embodiments, such kits do not include the materials and reagents for measuring the expression of a protein or RNA product of a biomarker of the invention that has been suggested by the prior art to be associated with bladder cancer. In other embodiments, such kits include the materials and reagents for measuring the expression of a protein or RNA product of ADAM 12, GSTP1 , FGFR3, MMP2, TWISTI , NID2, Vimentin, and/or p53, possibly in combination with a biomarker that has been suggested by the prior art to be associated with bladder cancer and any number of up to at least 1 , at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45 or more genes other than the combinatorial biomarkers of the invention.
The invention provides kits useful for determining whether a subject will be responsive to a therapy based upon detecting a protein or RNA product of ADAM 12, GSTP1, FGFR3, MMP2, TWISTI , NID2, Vimentin, and/or p53, possibly in combination with any number of up to at least 1 , at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20, at least 25, at least 30, at least 35, at least 40, at least 45, at least 50, all or any combination of the combinatorial biomarkers of the invention in a sample.
In a specific embodiment, such kits comprise materials and reagents that are necessary for measuring the expression of a RNA product of a biomarker of the invention. For example, a kit may comprise a microarray or RT-PCR kit. For nucleic acid microarray kits, the kits generally comprise probes attached to a solid support surface. The probes may be labelled with a detectable label. In a specific embodiment, the probes are specific for an exon(s), an intron(s), an exon junction(s), or an exon-intron junction(s)), of RNA products of ADAM12 possibly in combination with any number of up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention.
The microarray kits may comprise instructions for performing the assay and methods for interpreting and analyzing the data resulting from the performance of the assay. In a specific embodiment, the kits comprise instructions for diagnosing bladder cancer. The kits may also comprise hybridization reagents and/or reagents necessary for detecting a signal produced when a probe hybridizes to a target nucleic acid sequence. Generally, the materials and reagents for the microarray kits are in one or more containers. Each component of the kit is generally in its own a suitable container.
For RT-PCR kits, the kits generally comprise pre-selected primers specific for particular RNA products (e.g., an exon(s), an intron(s), an exon junction(s), and an exon-intron junction(s)) of ADAM 12, GSTPl, FGFR3, MMP2, TWISTl , NID2, Vimentin, and/or p53 possibly in combination with any number of up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention. The RT-PCR kits may also comprise enzymes suitable for reverse transcribing and/or amplifying nucleic acids (e.g., polymerases such as Taq), and deoxynucleotides and buffers needed for the reaction mixture for reverse transcription and amplification. The RT-PCR kits may also comprise probes specific for RNA products of ADAM 12, GSTPl , FGFR3, MMP2, TWISTl , NID2, VIMENTIN, and/or p53, and possibly any number of up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention. The probes may or may not be labelled with a detectable label (e.g., a fluorescent label). Each component of the RT-PCR kit is generally in its own suitable container. Thus, these kits generally comprise distinct containers suitable for each individual reagent, enzyme, primer and probe. Further, the RT-PCR kits may comprise instructions for performing the assay and methods for interpreting and analyzing the data resulting from the performance of the assay. In a specific embodiment, the kits contain instructions for diagnosing bladder cancer.
In a specific embodiment, the kit is a real-time RT-PCR kit. Such a kit may comprise a 96 well plate and reagents and materials necessary for e.g. SYBR Green detection. The kit may comprise reagents and materials so that beta-actin can be used to normalize the results. The kit may also comprise controls such as water, phosphate buffered saline, and phage MS2 RNA. Further, the kit may comprise instructions for performing the assay and methods for interpreting and analyzing the date resulting from the performance of the assay. In a specific embodiment, the instructions state that the level of a RNA product of ADAM 12, GSTPl , FGFR3, MMP2, TWISTl , NID2, Vimentin, and/or p53, and possibly any number of up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention should be examined at two concentrations that differ by, e.g., 5 fold to 10-fold.
For antibody based kits, the kit can comprise, for example: (1) a first antibody (which may or may not be attached to a solid support) which binds to ADAM 12, GSTPl , FGFR3, MMP2, TWIST1 , NID2, Vimentin, and/or p53 and any combinatorial protein of interest (e.g., a protein product of any number of up to 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, all or any combination of the combinatorial biomarkers of the invention); and, optionally, (2) a second, different antibody which binds to either the protein, or the first antibody and is conjugated to a detectable label (e.g., a fluorescent label, radioactive isotope or enzyme). The antibody-based kits may also comprise beads for conducting an immunoprecipitation. Each component of the antibody-based kits is generally in its own suitable container/Thus, these kits generally comprise distinct containers suitable for each antibody. Further, the antibody-based kits may comprise instructions for performing the assay and methods for interpreting and analyzing the data resulting from the performance of the assay.
In a specific embodiment, the kits contain instructions for diagnosing bladder cancer.
Incorporation by Reference
References and citations to other documents, such as patents, patent applications, patent publications, journals, books, papers, web contents, have been made throughout this disclosure. All such documents are hereby incorporated herein by reference in their entirety for all purposes.
Equivalents
The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting on the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.
EXAMPLES
EXAMPLE 1. Whole urine DNA recovery versus DNA recovery from cellular and cell-free portions of whole urine.
The amount of DNA recoverable from a whole urine sample was compared to the amount of DNA recoverable from a cellular (pellet) urine sample and a cell-free (supernantant) urine sample from an identical sample of whole urine. Sample Collection
1. 5 EDTA tubes (~ 8-10ml per tube) were received per individual sample. There were a total of 200 samples in this study.
2. 4 of the 5 EDTA tubes were pooled together. 16ml were aliquoted into two 50ml
conicals. One 50ml conical is labeled "Whole Urine".
3. The second 50ml conical is labeled "Pellet" (Cell-associated DNA) and is centrifuged at 1800g for 10 minutes. The supernatant is poured off into a new 50ml conical and is labeled "Free DNA".
4. The conical labeled "Pellet" is rinsed with 1ml of TE buffer, and is centrifuged at 3000g for 5 minutes. The supernatant is carefully removed with a pipet and discarded.
5. Samples were then stored at -80°C as well as the remaining EDTA tube.
DNA isolation
1. Whole Urine Analysis: 16ml of urine was prepared using the current Isopropanol
precipitation protocol (C-LAB-028).
2. Free DNA analysis: 16ml of whole urine was spun at 1800g for 10 minutes to remove intact cells. The supernatant from each was poured off and subjected to Isopropanol precipitation according to the current protocol.
3. Cell-associated DNA Analysis: The resulting cell pellet from the Free DNA was re- suspended in 16ml of TE buffer and subjected to Isopropanol precipitation according to the current protocol.
4. The DNA yield from each sample prepped was determined by Exon 7 Quantitation
(LAB-029) and expressed as total ng.
Results:
1. The amount of DNA required to detect 0.08% mutant DNA with 85% confidence is 22ng.
An additional 40ng are required for our hypermethylation assays. Therefore, a total of 62ng is required from each 16ml prep. The number of samples that meet this minimum requirement of total DNA yield is shown below for whole urine, cell-associated DNA and free DNA. Whole Urine Cell Associated DNA Free DNA
Number without 62ng 23 77 87
Total samples tested 200 200 200
Percent insufficient 1 1.5% 38.5% 43.5%
2. Out of the 200 samples tested, 156 (78%) had the highest yield when whole urine was prepped.
3. In these cases where the Whole Urine showed the highest DNA quantity, approximately half of the samples had a higher yield in the Free DNA component versus the Cell- Associated DNA. And was sample dependent. In samples with high DNA yield in whole urine, there was a trend towards the majority of the yield to be found in the cell associated component whereas in samples with lower yield, the majority of the DNA tended to be in the free DNA component.
4. In 145 out of 200 samples, the sum of the cell associated plus free DNA yields was lower than in whole urine, suggesting a loss in DNA yield with partitioning. In these samples, there was an average loss of 44% in yield.
From this data, we conclude that whole urine partitioning leads to loss in DNA yield and an increase in insufficient samples when working with a predetermined sample size. The data also suggests that whole urine analysis is more likely to provide meaningful numbers of DNA biomarkers in a clinical population.

Claims

1. A method for identifying a condition in a subject, comprising:
providing a first nucleic acid sample collected from a cell-free portion of a body fluid sample from a subject;
providing a second nucleic acid sample collected from a cellular portion of the body fluid sample;
assaying the first nucleic acid sample for a first marker; and
assaying the second nucleic acid sample for a second marker, wherein the presence of the first and second markers is indicative of a condition in the subject.
2. The method of claim 1 , wherein the presence of only the first marker or only the second marker is not sufficient to indicate the condition.
3. The method of claim 1 or 2 wherein the body fluid is selected from urine, whole blood, saliva, tears, sweat, sputum, cerebral spinal fluid, menstrual fluid, semen, and nipple aspirate.
4. The method of claim 3, wherein the body fluid is urine.
5. The method of any of claims 1 to 4, wherein the nucleic acid is DNA.
6. The method of any of claims 1 to 5, wherein the condition is cancer.
7. The method of any of claims 1 to 5, wherein the condition is relapse of cancer.
8. The method of claims 6 or 7, wherein the cancer is selected from bladder cancer, lung cancer, breast cancer, ovarian cancer, prostate cancer, uterine cancer, cervical cancer, pancreatic cancer, brain cancer, esophageal cancer, throat cancer, mouth cancer, lymphatic cancer, and leukemia.
9. The method of any of claims 1 to 8, wherein the first marker or the second markers comprise a mutation in a human gene.
10. The method of claim 9, wherein the mutation is in a gene selected from FGFR, TWIST, Vimentin, and NID.
1 1. The method of any of claims I to 8, wherein the first marker or the second markers comprise an epigenetic modification to the first or second nucleic acids.
12. The method of claim 1 1 , wherein the epigenetic modification is methylation or acetylation.
13. The method of claim 1 1, wherein the epigenetic modification is in a gene selected from FGFR, TWIST, Vimentin, and NID.
14. The method of claim 1 , further comprising:
providing the body fluid sample from the subject;
isolating the cell-free nucleic acid; and
isolating the cellular nucleic acid.
15. The method of claim 14, wherein isolating the cell-free nucleic acids and the cellular nucleic acid comprises centrifuging the body fluid sample.
16. The method of claim 15, wherein isolating the cell-free nucleic acids and the cellular nucleic acidare precipitated with an alcohol.
17. The method of claim 1 , further comprising directing a treatment for the condition.
18. The method of claim 1 , further comprising administering a therapeutic to the subject.
19. The method of any of claims 1 to 18, wherein the assay comprises a hybridization assay, nucleic acid sequencing, or quantitative PCR.
20. The method of claim 19, wherein nucleic acid sequencing comprises next generation sequencing.
21. A method for identifying a condition in a subject, comprising:
providing a nucleic acid sample comprising nucleic acids associated with a cellular component and nucleic acids associated with a cell-free component;
assaying both the nucleic acids associated with the cellular component and the nucleic acids associated with the cell-free component for a marker indicative of a condition in the subject.
22. The method of claim 21 wherein the body fluid is selected from urine, whole blood, saliva, tears, sweat, sputum, cerebral spinal fluid, menstrual fluid, semen, and nipple aspirate.
23. The method of claim 22, wherein the body fluid is urine.
24. The method of any of claims 21 to 23, wherein the nucleic acid is DNA.
25. The method of any of claims 21 to 24, wherein the condition is cancer.
26. The method of any of claims 1 to 5, wherein the condition is relapse of cancer.
27. The method of claim 21 , further comprising:
providing the body fluid sample from the subject;
isolating the nucleic acids associated with a cellular component; and
isolating the nucleic acids associated with a cell-free component.
28. The method of claim 27, wherein the nucleic acids associated with a cellular component and the nucleic acids associated with a cell-free component are isolated in the same step.
29. The method of claim 27, wherein the nucleic acids associated with the cellular component or the cell-free component are precipitated with an alcohol.
30. The method of claim 27, wherein the body fluid sample is not centrifuged.
PCT/US2014/036751 2013-05-06 2014-05-05 Diagnostic assay combining cellular and cell free nucleic acid WO2014182598A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361820061P 2013-05-06 2013-05-06
US61/820,061 2013-05-06

Publications (1)

Publication Number Publication Date
WO2014182598A1 true WO2014182598A1 (en) 2014-11-13

Family

ID=51867659

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/036751 WO2014182598A1 (en) 2013-05-06 2014-05-05 Diagnostic assay combining cellular and cell free nucleic acid

Country Status (1)

Country Link
WO (1) WO2014182598A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104745700A (en) * 2015-03-27 2015-07-01 南京医科大学 Esophageal-cancer-related methylated biomarker and application thereof
CN105785004A (en) * 2016-03-31 2016-07-20 中国医学科学院北京协和医院 Application of cell cycle division associated protein 2 to diagnosis or prognosis of pancreatic cancer
WO2019126249A1 (en) * 2017-12-20 2019-06-27 Laboratory Corporation Of America Holdings Compositions and methods to detect head and neck cancer
JP2019522463A (en) * 2016-05-16 2019-08-15 ナノストリング テクノロジーズ,インコーポレイティド Method for detecting a target nucleic acid in a sample
CN114150071A (en) * 2020-09-08 2022-03-08 河南农业大学 Application of SNP molecular marker of chicken TRIM13 gene in chicken growth and slaughter trait improvement breeding and breeding method
US11279969B2 (en) 2016-11-21 2022-03-22 Nanostring Technologies, Inc. Chemical compositions and methods of using same
US11549139B2 (en) 2018-05-14 2023-01-10 Nanostring Technologies, Inc. Chemical compositions and methods of using same
WO2023147568A3 (en) * 2022-01-28 2023-08-31 Flagship Pioneering Innovations Vi, Llc Compositions and methods for making and using an immortalized library

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005081867A2 (en) * 2004-02-20 2005-09-09 The Regents Of The University Of C Alifornia Salivary mrna profiling, biomarkers, and related methods and kits of parts
WO2008155549A2 (en) * 2007-06-19 2008-12-24 Oncomethylome Sciences Sa Improved urine sample collecting and processing
US20120142001A1 (en) * 2008-02-01 2012-06-07 Exosome Diagnostics, Inc. Method for isolation of nucleic acid containing particles and extraction of nucleic acids therefrom

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005081867A2 (en) * 2004-02-20 2005-09-09 The Regents Of The University Of C Alifornia Salivary mrna profiling, biomarkers, and related methods and kits of parts
WO2008155549A2 (en) * 2007-06-19 2008-12-24 Oncomethylome Sciences Sa Improved urine sample collecting and processing
US20120142001A1 (en) * 2008-02-01 2012-06-07 Exosome Diagnostics, Inc. Method for isolation of nucleic acid containing particles and extraction of nucleic acids therefrom

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
KOHLER ET AL.: "Cell -free DNA in the circulation as a potential cancer biomarker", ANTICANCER RESEARCH, vol. 31, no. 8, 2011, pages 2623 - 2628 *
NILSSON ET AL.: "Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer", BRITISH JOURNAL OF CANCER, vol. 100, no. 10, 2009, pages 1603 - 1607 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104745700A (en) * 2015-03-27 2015-07-01 南京医科大学 Esophageal-cancer-related methylated biomarker and application thereof
CN105785004A (en) * 2016-03-31 2016-07-20 中国医学科学院北京协和医院 Application of cell cycle division associated protein 2 to diagnosis or prognosis of pancreatic cancer
JP2019522463A (en) * 2016-05-16 2019-08-15 ナノストリング テクノロジーズ,インコーポレイティド Method for detecting a target nucleic acid in a sample
US11279969B2 (en) 2016-11-21 2022-03-22 Nanostring Technologies, Inc. Chemical compositions and methods of using same
US11821026B2 (en) 2016-11-21 2023-11-21 Nanostring Technologies, Inc. Chemical compositions and methods of using same
WO2019126249A1 (en) * 2017-12-20 2019-06-27 Laboratory Corporation Of America Holdings Compositions and methods to detect head and neck cancer
US11933784B2 (en) 2017-12-20 2024-03-19 Laboratory Corporation Of America Holdings Compositions and methods to detect head and neck cancer
US11549139B2 (en) 2018-05-14 2023-01-10 Nanostring Technologies, Inc. Chemical compositions and methods of using same
CN114150071A (en) * 2020-09-08 2022-03-08 河南农业大学 Application of SNP molecular marker of chicken TRIM13 gene in chicken growth and slaughter trait improvement breeding and breeding method
CN114150071B (en) * 2020-09-08 2023-07-25 河南农业大学 Application of chicken TRIM13 gene SNP molecular marker in chicken growth and slaughter trait improvement breeding and breeding method
WO2023147568A3 (en) * 2022-01-28 2023-08-31 Flagship Pioneering Innovations Vi, Llc Compositions and methods for making and using an immortalized library

Similar Documents

Publication Publication Date Title
US20120252020A1 (en) Screening Assay for Bladder Cancer
WO2014182598A1 (en) Diagnostic assay combining cellular and cell free nucleic acid
ES2616800T3 (en) Obtaining gene expression profile in biopsied tumor tissues
US8841095B2 (en) Detection of nucleic acids and proteins
Zonta et al. Assessment of DNA integrity, applications for cancer research
US20150126400A1 (en) Molecular diagnostic screening assay
WO2013173485A1 (en) Detection of bladder cancers
EA037995B1 (en) Biomarker panel for the detection of cancer
JP2019162102A (en) System and method of detecting rnas altered by cancer in peripheral blood
US20150079033A1 (en) Detection of bladder cancer and recurrent bladder cancer
CN114269916A (en) Device and method for sample analysis
Duployez et al. Minimal residual disease monitoring in t (8; 21) acute myeloid leukemia based on RUNX1‐RUNX1T1 fusion quantification on genomic DNA
CN114945687A (en) Characterization of methylated DNA, RNA and protein in a subject suspected of having a lung neoplasia
US8852893B2 (en) Detection of nucleic acids and proteins
Nazir Medical diagnostic value of digital PCR (dPCR): A systematic review
KR101704533B1 (en) Err as the biomaker to liver cancer and use thereof
US20120244536A1 (en) Detection of Bladder Cancer Recurrence
WO2013173478A1 (en) Screening assay for bladder cancer
US8377657B1 (en) Primers for analyzing methylated sequences and methods of use thereof
EP3301186B1 (en) Method, sequences, compositions and kit for detection of mutationen in the promoter of the gene htert
US20130309667A1 (en) Primers for analyzing methylated sequences and methods of use thereof
CN108277274A (en) Composition and application thereof for differentiating pancreatic cancer and chronic pancreatitis
WO2013173480A1 (en) Detection of bladder cancer recurrence
WO2013173476A1 (en) Detection of cancer
RU2193069C2 (en) Method of detection of mutations of deoxyribonucleic acids using restrictases and kit for assay carrying out

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: 14794108

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14794108

Country of ref document: EP

Kind code of ref document: A1