GB2327497A - Diagnostic assay for cancer - Google Patents

Diagnostic assay for cancer Download PDF

Info

Publication number
GB2327497A
GB2327497A GB9815224A GB9815224A GB2327497A GB 2327497 A GB2327497 A GB 2327497A GB 9815224 A GB9815224 A GB 9815224A GB 9815224 A GB9815224 A GB 9815224A GB 2327497 A GB2327497 A GB 2327497A
Authority
GB
United Kingdom
Prior art keywords
ras
diagnostic
arms
primer
dna
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
GB9815224A
Other versions
GB2327497B (en
GB9815224D0 (en
Inventor
Richard Mark Ferrie
Gillian Ellison
Kay Callaghan
Jayne Catherine Fox
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Syngenta Ltd
Original Assignee
Zeneca Ltd
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 Zeneca Ltd filed Critical Zeneca Ltd
Publication of GB9815224D0 publication Critical patent/GB9815224D0/en
Publication of GB2327497A publication Critical patent/GB2327497A/en
Application granted granted Critical
Publication of GB2327497B publication Critical patent/GB2327497B/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/16Primer sets for multiplex assays

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)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Biochemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

A diagnostic assay for the detection of K-ras mutations in cancer. The method comprises contacting a test sample of nucleic acid with a diagnostic primer for a K-ras mutation in the presence of appropriate nucleotide triphosphates and an agent for polymerisation, such that the diagnostic primer is extended only when a K-ras mutation is present in the sample; and detecting the presence or absence of a diagnostic primer extension product. Diagnostic primers for seven K-ras point mutations are provided. Also included is a diagnostic kit in which one or more diagnostic primers are conveniently packaged with appropriate nucleotide triphosphates, polymerase, buffer and instructions for use. Amplification from the diagnostic primer may be by ARMS or ALEX techniques, and may be monitored by real time detection of product.

Description

ASSAY This invention relates to a diagnostic method for the detection of K-ras mutations in cancer, particularly colorectal cancer, using the amplification refractory mutation system, (ARMS).
The invention also relates to mutation specific primers for use in the method and to diagnostic kits containing these primers.
DescriDtion Colorectal cancer is the second most common cause of cancer mortality in the US and is a significant cause of morbidity and mortality world-wide. The American Cancer Society estimated that in 1995 more than 130,000 new cases of colorectal cancer (CRC) would be diagnosed in the US and that there would be 55,300 deaths from the disease. The incidence is higher in men than in women (60.4 versus 40.9 per 100,000 per year). Agespecific incidence and mortality rates show that most cases are diagnosed after 50 years of age. Most patients (65%) present with advanced disease with about 60% of patients having regional or distant metastases at the time of diagnosis. The estimated 5-year survival is 91% in persons with localised disease, 60% in persons with regional spread, and only 6% in those with distant metastases. An individual's lifetime risk of dying of colorectal cancer in the US has been estimated to be 2.6%. CRC onset and its progression have been studied extensively at the molecular and genetic levels and there is a commonly accepted model relating tumour status, classified according to Duke's stage, to specific changes in DNA.
It has recently been concluded that reliable CRC screening procedures require development and that additional research to identify mutated genes in clinical samples is needed. The United States Preventive Services Task Force also recommends screening for CRC for all persons aged 50 and older with annual faecal occult blood testing (FOBT) or sigmoidoscopy or both. The reported sensitivity and specificity of FOBT for detecting colorectal cancer in asymptomatic persons are 26-92% and 90-99%, respectively (usually based on two samples from three different stool specimens). However, the ingestion of foods containing peroxidases, and gastric irritants such as salicylates and other antiinflammatory agents, can produce false-positive test results for neoplasia. Nonneoplastic conditions, such as haemorrhoids, diverticulosis, and peptic ulcers, can also cause gastrointestinal bleeding that would give rise to false-positive test results. FOBT can also miss small adenomas and colorectal malignancies that bleed intennittently or not at all. Other causes of false-negative results include heterogeneous distribution of blood in faeces, ascorbic acid and other antioxidants that interfere with test reagents, and extended delay before testing stool samples.
As a result, when FOBT is performed on asymptomatic persons, the majority of positive reactions are falsely positive for neoplasia. The reported positive predictive value among asymptomatic persons over age 50 is only about 2-11% for carcinoma and 20-30% for adenomas. Assuming a false-positive rate of 1-4%, a person who receives annual FOBT from age 50 to age 75 has an estimated 45% probability of receiving a false-positive result. This large proportion of false-positive results is an important concern because of the discomfort, cost, and occasional complications associated with follow-up diagnostic tests, such as barium enema and colonoscopy. Other tests have been proposed to improve the accuracy of screening for faecal occult blood. Current evidence is equivocal as to whether "HemoQuant" (SmithKline Diagnostics, Sunnyvale, CA), a quantitative measurement of haemoglobin in the stool, has better sensitivity or specificity than does qualitative FOBT.
Sigmoidoscopic screening in asymptomatic persons detects 14 cancers per 1,000 examinations. However, the sensitivity and diagnostic yield of sigmoidoscopy screening varies with the type of instrument. Since only 30% of colorectal cancers occur in the distal 20 cm of bowel, and less than half occur in or distal to the sigmoid colon, the length of the sigmoidoscope has a direct effect on case detection. The rigid sigmoidoscope, which has an average depth of insertion of about 20 cm and allows examination to just above the rectosigmoid junction, can detect only about 25-30% of colorectal cancers. The 35 cm flexible sigmoidoscope, however, can visualise about 50-75% of the sigmoid colon. The longer 60 cm instruments have an average depth of insertion of 40-50 cm, reaching the proximal end of the sigmoid colon in 80% of examinations with the capability of detecting 40-65% of colorectal cancers.
Other potential screening tests for colorectal cancer include colonoscopy and barium enema, which appear to have comparable accuracy. About 95% of colorectal cancers are within reach of the colonoscope, and the examination has an estimated 75-95% sensitivity in detecting lesions within its reach. The introduction of flexible fibre-optic instruments has made sigmoidoscopy more acceptable to patients, however, the procedure is expensive and remains uncomfortable and embarrassing, and therefore many patients may be reluctant to agree to this test. Colonoscopy, which requires sedation and often involves the use of a hospital suite, is more expensive than other screening tests and has a higher risk of anaesthetic and procedural complications.
There are tumour markers that are detected by immunodiagnostic techniques. For example, carcinoembryonic antigen (CEA), an oncofetal glycoprotein antigen with unknown function. Elevated serum levels of CEA correlate with Duke's stage of CRC. Elevated CEA levels are also evident in breast, liver, lung, pancreatic and gastric neoplasms. The limitations of tests based on antibodies raised to such tumour-associated antigens are related to both specificity and sensitivity.
Although there are many examples of nucleic acid changes having potential as tumour markers, their value as clinical tools in cancer diagnosis, staging or even screening, needs to be demonstrated and two important criteria must be met. Firstly, nucleic acids of adequate yield and quality must be extracted from the clinical material; secondly, robust and accurate methods of analysis are required. For reliable tumour genotyping to be useful in disease staging any test has to be adequately validated and there should be demonstrable benefits over current methods.
A number of studies have examined the association of CRC with mutations in the oncogene, K-ras. However, there have been significant differences in the reported frequencies of K-ras mutations in CRC. Inconsistencies between studies could be due to one or more of several factors. These include the number of tumours investigated, the methods used and the number of individual point mutations tested for. It is therefore difficult to state the true number of CRCs which contain K-ras mutations. Some associations between K-ras genotype and phenotype have been proposed. For instance, K-ras genotypes vary by amino acid substitution and have been categorised into different types. In a recent study, K-ras mutations have been shown to be associated with increased risk of relapse and death, with some mutations, such as the codon 12 glycine to valine change being more aggressive than others (Jervoise, H. et al., J.Nat.Cancer Inset. 90 675-684,1998).
In the present invention we have now devised novel diagnostic methods for the detection of K-ras mutations based on ARMS. Validated tests for seven K-ras point mutations have been developed and the tests have been applied in a thorough investigation of the incidence of the mutations in tumours from a large assembly of CRC patients.
According to a first aspect of the invention we now provide a diagnostic method for the detection of K-ras mutations in cancer, which method comprises contacting a test sample of nucleic acid with a diagnostic primer for a K-ras mutation in the presence of appropriate nucleotide triphosphates and an agent for polymerisation, such that the diagnostic primer is extended only when a K-ras mutation is present in the sample; and detecting the presence or absence of a K-ras mutation by reference to the presence or absence of a diagnostic primer extension product.
Each of the diagnostic primers detects one of the following K-ras mutations: Table 1:
PRIMBR MUTAON DETECmD NUMBER 1 K-ras codon 12 GGToAGT (glycine # serine) 2 K-ras codon 12 GGToCGT (glycine o arginine) 3 K-ras codon 12 GGT#TGT (glycine < cysteine) 4 K-ras codon 12 GGToGAT (glycine e aspartic acid) 5 K-ras codon 12 GGTeGCT (glycine < alanine) 6 K-ras codon 12 GGT#GTT (glycine e valine) 7 K-ras codon 13 GGCoGAC (glycine e aspartic acid) It is believed that the diagnostic methods of the invention may be used to detect a range of cancers, for example non-small cell lung cancer, pancreatic cancer and, in particular, colorectal cancer.
The test sample of nucleic acid is conveniently a sample of blood, faeces, sputum, colonic lavage, bronchial lavage or other body fluid, or tissue obtained from an individual.
The individual is conveniently human, preferably Homo sapiens. It will be appreciated that the test sample may equally be a nucleic acid sequence corresponding to the sequence in the test sample. That is to say that all or a part of the region in the sample nucleic acid may firstly be amplified using any convenient technique such as PCR before use in the method of the invention.
Any convenient enzyme for polymerisation may be used provided that it does not affect the ability of the DNA polymerase to discriminate between normal and mutant template sequences to any significant extent. Examples of convenient enzymes include thermostable enzymes which have no significant 3'-5' exonuclease activity, for example Taq DNA polymerase, particularly "Ampli Taq Gold"M DNA polymerase (PE Applied Biosystems), Stoffel fragment, or other appropriately N-terminal deleted modifications of Taq or Tth (Thermus thermophilus) DNA polymerases.
We have now devised primers for the above K-ras point mutations which have been shown to detect the specific mutations reliably and robustly. Therefore in a further aspect of the invention we provide diagnostic primers comprising the sequences given below and derivatives thereof wherein 6-8 of the nucleotides at the 3' end are identical to the sequences given below and wherein up to 10, such as up to 8, 6, 4, 2, 1, of the remaining nucleotides may be varied without significantly affecting the properties of the diagnostic primer.
Conveniently, the sequence of the diagnostic primer is exactly as written below.
Table 2:
PRIMER MUTATION PRIMER SEQUENCE NUMBER DETECTED 1 K12 Serine CTGAATATAAACTTGTGGTAGTTGGAGCCA 2 @ K12 Arginine CTGAATATAAACTTGTGGTAGTTGGAGCCC 3 K12 Cysteine CTGAATATAAACTTGTGGTAGTTGGAGCAT 4 K12 Aspartic acid TATCGTCAAGGCACTCTTGCCTACGCCTT 5 K12 Alanine TATCGTCAAGGCACTCTTGCCTACGCCTG 6 K 1 2 Valine TATCGTCkAGGCACTCTTGCCTACGCCA 7 1 K13 Aspartic acid CGTGTATCGTCAAGGCACTCTTGCCTACCT The primers may be manufactured using any convenient method of synthesis.
Examples of such methods may be found in standard textbooks, for example "Protocols For Oligonucleotides And Analogues: Synthesis And Properties;" Methods In Molecular Biology Series; Volume 20; Ed. Sudhir Agrawal, Humana ISBN: 0-89603-247-7; 1993; 1st Edition.
It will be appreciated that any of the above diagnostic methods may, if appropriate, also be configured so that extension of the diagnostic primer indicates the absence of the K-ras mutation.
In many situations, it will be convenient to use a diagnostic primer of the invention with a further amplification primer in one or more cycles of PCR amplification. A convenient example of this aspect is set out in our European patent number EP-B 1-0332435. The further amplification primer is either a forward or a reverse common primer. The forward primer: GTACTGGTGGAGTATTTGATAGTGTATTAACC is conveniently used in combination with one or more of diagnostic primers 4, 5, 6 & 7 and the reverse primer: CTCATGAAAATGGTCAGAGAAACCTTTATC is conveniently used in combination with one or more of diagnostic primers 1, 2, & 3.
Any convenient control primer may be used. We have selected control primers from two unrelated regions of the genome, namely part of exon 4 of the cystic fibrosis transmembrane regulator gene, and part of exon II and intron m of the human a1-antitrypsin gene.
It will be appreciated that further useful diagnostic tests will be provided by combining 2 or more diagnostic primers for the 7 loci indicated above. Any appropriate combination of the above primers may be used. Particular combinations include primers 1+2, primers 1+2+3, primers 4+5, primers 4+5+6, and primers 4+5+6+7. Each of these combinations will also include the addition of the appropriate common primer.
The diagnostic methods of the invention as outlined above may be conveniently effected in one or more reaction vessels. Where more than one diagnostic mutation is to be assayed the diagnostic primer (and corresponding amplification primer) are provided in individual tubes i.e. one tube per mutation. Alternatively, the reactions may be multiplexed, that is to say that all the diagnostic primers and amplification primers are in one tube (see EP B 1-0332435).
A variety of methods may be used to detect the presence or absence of diagnostic primer extension products and/or amplification products. These will be apparent to the person skilled in the art of nucleic aid detection procedures. Preferred methods avoid the need for radiolabelled reagents. Particular detection methods include "Taqman" product detection, for example as described in patent numbers US-A-5487972 & US-A-5210015; "Molecular Beacons" product detection, outlined in patent number WO-95/13399 and surface enhanced ramon resonance spectroscopy (SERRS), outlined in our patent application WO 97/05280.
Further preferred detection methods include ARMS linear extension (ALEX) and PCR with ALEX as described in Example 4 of this application. Conveniently, real-time detection is employed.
One or more of the diagnostic primers of the invention may be conveniently packaged with instructions for use in the method of the invention and appropriate packaging and sold as a kit. The kits will conveniently include one or more of the following: appropriate nucleotide triphosphates, for example dATP, dCTP, dGTP, dTTP, a suitable polymerase as previously described, and a buffer solution.
The invention will now be illustrated but not lirnited by reference to the following Example, Tables and Figures.
Figure Legends Legend to Figure 1.
The K-ras genomic DNA sequence; wild-type codons 12 and 13 are shown in bold type. Filled arrows (+ and +) identify the 3' end of the ARMS primers designed to amplify individual K-ras mutated sequences. Open arrows ( and ) identify the 3' ends of the flanking intron. The ARMS primers were designed to detect point mutations of K-ras codon 12 glycine (GGT) to arginine (CGT), cysteine (TGT), serine (AGT), valine (GTT), aspartic acid (GAT) and alanine (GCT) and point mutation of codon 13 glycine (GGC) to aspartic acid (GAC). The direction of the arrow heads indicates sense (+ and ) and antisense ( and ) primers. ARMS reaction products derived from the intron-specific primer cpA () and any of the antisense ARMS primers (+) are 158 base-pairs (codon 12 mutations) or 161 base-pairs (codon 13 mutation). ARMS amplicons derived from the intron-specific primer cpB () and any of the sense ARMS primers (+) are 190 base-pairs.
Legend to Figure 2 Development of the K12 Asp (panel A), K12 Cys (panel B), K13 Asp (panel C) and K12 Val (panel D) ARMS tests. In each case lanes 1 and 2 are tests on tumour DNA; lane 3, 103 genome equivalents wild-type DNA; lane 4, 103 genome equivalents mutant (cell line) DNA; lane 5, 103 genome equivalents wild-type DNA plus 102 genome equivalents mutant DNA; lane 6, no DNA.
Legend to Figure 3 Example of a typical ARMS analysis on tumour DNA. DNA from selected Dukes' C tumours was tested using the K12 Ala ARMS test. Lane 1, x174/Haem size markers; lanes 2-14, tumours 1305, 5, 6, 13, 20, 21, 23, 39, 121, 122, 135, 137 and 142 respectively; lane 15, 105 genome equivalents SW116 cell line DNA; lane 16, no DNA.
Legend to Figure 4 Three-primer PCR with cloned K-ras exon I amplicons. Lanes 1 and 48, x174/Haem size markers; lanes 246, PCR aliquots from reactions carried out with the M13 forward and reverse primers and the ARMS primer that gave the preliminary ARMS result.
High molecular weight bands signify the presence of cloned insert; low molecular weight bands signify that no insert is present in the clone. Intermediate molecular weight bands are derived from the respective ARMS primer and one or other of the M13 primers, the slightly different sizes of these bands signify the use of either a sense or an antisense ARMS primer. Lane 47, no DNA.
Legend to Figure 5 Control data graphs showing homogeneous detection of K-ras mutant sequences using ARMS coupled with amplicon detection by Molecular Beacons. Data points were subjected to regression analysis with 98% confidence limits. Data for each K-ras mutation is shown as follows: Figure 5a K-ras codon 13 Gly to Asp Figure 5b K-ras codon 12 Gly to Asp Figure 5c K-ras codon 12 Gly to Val Figure sod K-ras codon 12 Gly to Ala Figure Se K-ras codon 12 Gly to Ser Figure Sf K-ras codon 12 Gly to Arg Figure 52 K-ras codon 12 Gly to Cys Examnle 1 In Example 1 we report a study of the incidence of K-ras mutations in colorectal cancer using 301 DNA samples extracted from a colorectal tumour bank. In this study mutations within codons 12 and 13 of the K-ras oncogene were investigated using ARMS (Newton C.R. et al., Nucl. Acids Res., 17: 2503-2516, 1989). Direct DNA sequencing (Newton C.R. et al., Nucl. Acids Res., 16: 8233-8243, 1988), and sequencing of cloned amplicons were then performed to assess the ARMS test results. In nearly half of the cases the direct sequencing result detected K-ras wild-type sequence only, this was in contrast to the ARMS findings where mutations in codons 12 and 13 were detected. When K-ras exon I amplicons were cloned into E.coli, the ARMS result was consistently in accord with the sequence of the cloned material with the exception of one tumour which harbored a mutation of five consecutive nucleotides that was detected by three of the ARMS primers. Our data show that ARMS is a sensitive test for detecting under-represented nucleic acid sequences. We also demonstrate that the technique is ideally suited to the detection of tumour DNA markers supplying genotype information specific to pre-diagnosed tumours.
Materials And Methods (1) DNA Extraction From Tumour Bank Samples.
DNA was extracted from 301 frozen tissue samples. Positive selection of samples comprised the exclusion of adenomas and tumours from familial adenomatis polyposis patients.
Altogether, the samples comprised colorectal lesions excised between January 1985 and January 1995, 6 of which were adenomas. Of the remaining cancers 31 were classified as Dukes' stage A, 135 as Dukes' B and 129 as Dukes' C (Dukes C.E., J. Pathol. Bacteriol., 35: 323-332, 1932).
Each frozen specimen was sectioned by cryostat, 5llm was taken for haematoxylin and eosin staining and 3 or 4 parallel claim sections were transferred to sterile tubes and stored at -70 C. Fresh blades were used for each sample.
DNA extraction comprised thawing sections on ice followed by the addition of sufficient sterile lysis buffer [lOmM Tris-HC1 (pH 7.5), 20% sodium dodecyl sulfate, 50pg/ml proteinase K] to saturate the material. After an overnight digestion at 370C a standard phenol/chloroform purification and ethanol precipitation was carried out (Sambrook J., Fritsch E.F. & Maniatis T.
(eds) Molecular Cloning: A Laboratory Manual. 2nd edition Cold Spring Harbor Laboratory Press (1989)). The resulting DNA was resuspended in 200Cl1 10mM Tris-HCl (pH 7.5). DNA samples were then stored at -700C prior to quantification and K-ras mutation analysis.
(2) K-ras Mutation ARMS Tests.
Individual ARMS tests were developed to detect specific point mutations in the Kras oncogene. The 3'-terminal base of each of seven ARMS oligonucleotide primers was complementary to one of the common mutations of codons 12 or 13 of the K-ras oncogene occurring in CRCs (Breivik J. et al., Br. J., Cancer, 69: 367-371, 1994), (Fig. 1). In addition to the ARMS primers, a common primer complementary to K-ras intron sequence was included (Fig. 1). ARMS and common primer sequences are shown in Table 3.
Two other primer pairs were also present in each test to give amplification control products. Their sequences were: 5'-TATATGTGCCATGGGGCCTGTGCAAGGAAG-3' and 5'-CTCCTACACCCAGCCATTITTGGC-3' which amplify part of exon four of the cystic fibrosis transmembrane conductance regulator (CFTR) gene (Riordan J.R. et al., Science, 245:10661073, 1989) and 5'-GGGCCTCAGTCCCAACATGGCTAAGAGGTG-3' and 5'-CCCACCTTCCCCTCTCTCCAGGCAAATGGG-3' which amplify a part of each of exon II and intron m of the human &alpha;1-antitrypsin gene (Newton C.R. et al., Nucl. Acids Res., 16: 8233-8243, 1988).
Normal human DNA was extracted from the blood of healthy volunteers (Ferrie R.M. et al., Am. J. Hum. Genet., 51: 251-262, 1992). K-ras mutated DNA samples were extracted from tumour derived cell lines as shown in Table 4. The K-ras mutation for each cell line DNA was confirmed by direct DNA sequencing as described below for tumour derived DNA samples. The cell line DNAs were then used to define amplification conditions that conferred specificity to each ARMS reaction when the following criteria were applied.
First, after the DNA amplification reaction and agarose gel electrophoresis in the presence of 0.511g/ml ethidium bromide (Sambrook J., Fritsch E.F. & Maniatis T., op.cit.), the test should give a visible ARMS band only when either 102 genome equivalents of the appropriate mutant DNA with 105 equivalents of normal DNA is combined or when 102 genome equivalents of mutant DNA alone is tested. A genome equivalent meaning here the amount of genomic DNA per cell.
In addition, there should be no visible ARMS product from any primer when 105 genome equivalents of normal DNA is tested in isolation.
The annealing temperature and number of cycles used for each ARMS reaction is shown in Table 5. All amplification reactions were performed applying the commonly accepted precautions for avoiding carry-over contamination (Kwok S. & Higuchi R., Nature, 339: 237238, 1989) The amount of DNA extracted from each tumour sample was measured by fluorescence following intercalation of the Hoechst dye 33258 (Riley J. et al., Nucl. Acids Res., 17: 8383, 1989).
ARMS reactions containing these DNAs (1 p1 each) were performed in 50111 buffer comprising l0mM Tris-HCl, (pH 8.3), 1.2mM MgCl2, 50mM KCl, 0.01% gelatin and dNTPs (100mM each). The reactions also contained mutation-specific and the appropriate intronspecific primers (1pM each) as shown in Fig 1. The CFTR gene amplimers were 0.075pM each and the a1-antitrypsin primers were 0.025pM each.
Hot-start PCR (D' Aquila R.T. et al., Nucl. Acids Res., 19: 3749, 1991; Chou Q. et al., Nucl. Acids Res., 20: 1717-1723, 1992) was performed throughout by adding a layer of white mineral oil and heating the samples at 94"C for 5 minutes before adding Taq DNA polymerase (1 unit). Thermal cycling comprised of 35 or 36 cycles (Table 5) of 94"C, 1 minute denaturation; 58 -63 C, 1 minute annealing (Table 5); 72 C, 1 minute extension. This was followed by a final incubation at 72 C for 10 minutes.
Any sample which failed to amplify, identified by the absence of control bands, was re-tested until data for all tumour samples with all 7 tests was complete.
(3) K-ras ARMS Test Validation: Direct Sequencing Of Tumour Derived DNA.
Tumour derived DNA (2il) was amplified in 50p1 reactions which comprised 10mM Tris-HCl (pH 8.3), 100mM tetramethylammonium chloride, 3mM MgCl2, 0.05% Tween-20, 0.05% Nonidet NP40, dNTPs (200 M each) and 2.5 units of Taq DNA polymerase.
Each reaction also contained the forward primer 5'-CTGGATCTAGACTCATGAAAATGGTCAGAGAAACCTTTATC-3' and the reverse primer 5'-CCTCGGAATTCGTACTGGTGGAGTATTTGATAGTGTATTAACC-3' (500nM each) which generate an amplicon from exon I of the K-ras oncogene with flanking XbaI and EcoRI restriction enzyme recognition sites.
Reactions were overlaid with mineral oil (50 1ll) and amplified over 35 cycles of 94 C, 60 C 72 C (1 minute each). After electrophoresis through a 2% metaphor agarose gel (FMC Bioproducts), the exon I bands were excised and purified using a "Wizard" DNA purification kit (Promega). Typical yields were 1-5pg in 50il.
The purified products were sequenced by direct incorporation of [a35S]dATP (Amersham) using a modified version (Green P.M. et al., The EMBO Journal, 8: 1067-1072, 1989) of the "Sequenase 2.0" DNA sequencing kit (Amersham). Annealing mixtures also contained template DNA (6 l), sequencing primer (1il, 500ng) and dimethylsulphoxide (1p1), (Sigma). Each labeling reaction was supplemented with 0.2units DNA polymerase I, Klenow fragment (labeling grade, Boehringer Mannheim) (Redston M.S. & Kern S.E., Biotechniques, 17: 286-288, 1994). Sequencing reactions were run on 6% polyacrylamide gels which were subsequently dried and autoradiographed.
(4) K-ras ARMS Test Validation: Cloning and Sequencing of Tumour DNAs.
An aliquot of each amplicon prepared for direct DNA sequencing was also ligated into the vector pGEM-T (Promega) at 17"C overnight. A 2cell aliquot from each ligation mixture was used to transform competent E.coli JM109 cells (Promega). These were plated and blue/white screened according to the supplier's instructions. White colonies were transferred to 10 Crl of sterile distilled water.
PCR using the conditions described above, but for 25 cycles, was carried out to test simultaneously for the presence of an insert and the K-ras mutation status of any insert.
The amplimers in each reaction were the M13 5'-GTTTTCCCAGTCACGAC-3' (forward), 5' CAGGAAACAGCTATGAC-3' (reverse) primers and the ARMS primer that initially identified the mutation. Amplification products were then visualised on a 3% agarose gel.
Clones with inserts detected by the PCR screen were transferred to lml of LB broth and grown overnight at 37"C. An aliquot (100X) from each was inoculated into a further lml broth and grown for 34 h at 370C. Ten pfu/cell of M13KO7 helper phage were then added to each culture. After lh at room temperature LB broth (9
(2) ARMS Test Development.
Thermal cycling and primer annealing conditions were determined empirically having imposed the specificity criteria described above, these are shown in Table 5. An example showing the specificity of each of four of the tests is shown in Figure 2.
(3) DNA Yield From Tumour Extracts.
The maximum and minimum DNA yields were 124.5clog (622.5 ng/W) and l.5Rg (7.5 ng/l) respectively, approximating to between 1.25x105 and l.5x103 human diploid genome equivalents/pl.
Six of the Dukes' C samples failed to give sufficient DNA to reach the threshold of detection but this did not preclude ARMS analyses of these samples. In general, a lower yield of DNA was associated with the extracts from Dukes' C classified tumours. The mean DNA yield was 25.6Rg (128.0 ng/X), approximately 2.5x104 genome equivalents/ after discounting the six samples that failed to give measurable quantities of DNA extract.
(4) ARMS Tests And Direct Sequence.
A typical ARMS result is shown in Figure 3. A summary of the K-ras mutations detected using ARMS, the Dukes' stage of the tumour and the direct DNA sequencing result for 30 of the tumour DNAs examined is shown in Table 6.
(5) Ouantitative Analysis of K-ras Mutations.
Three primer PCR results used to classify clones from a selection of tumour DNAs are shown in Figure 4. Table 7 provides an analysis of the ARMS data, direct DNA sequencing and clone analyses from equivalent samples.
(6) K-ras Mutational Analysis of the 301 Tumour Bank Samples.
Since the majority of CRCs are classified as either Dukes' B or C at the time of surgery a relatively smaller number of Dukes' A (31 in total, 10.3%) cancers was analysed.
Adenomas comprised 6 in total (2%) for the same reason and also because of the deliberate selection against these tumours. For the 295 non-adenoma tumours, there were more from male than female patients (183 male, 118 female), 36% had K-ras mutations and there were relatively higher numbers of severe disease patients 31, 135 and 129 Dukes' stages A, B and C respectively. There were relatively more severe male than female patients (male:female ratio for each Dukes' stage is; A, 15: 16; B, 74:61; C, 90:39; p < 0.05). For the proportion of K-ras mutations, there was evidence that this was less (p < 0.05) for males (31.7%) than females (43.2%). There was no evidence that the proportions differed across Dukes' stages either for the sexes separately or combined (combined proportions, Dukes' A, 13:18,42%; Dukes' B,47:88, 35%; and Dukes' C 48:81, 37%; p > 0.05). Table 6 shows the results on which the Kras codons 12 and 13 mutational analysis was made and the relative K-ras mutation frequencies.
The aims of our study were to develop validated tests for seven K-ras point mutations and to apply them in a thorough investigation of the incidence of the mutations in tumours from a large assembly of CRC patients. As part of the ARMS test validation process we employed the sequencing strategy described in Materials and Methods. Our initial approach to directly sequence PCR amplicons verified the ARMS result in approximately half of the tumour DNA samples investigated. One possible explanation for this could be that the ARMS tests failed to discriminate mutated from normal sequences. However, the tumours were not microdissected, but a large proportion of tumour cells relative to normal tissue was identified by histology. Assuming that the tumour cells were monoclonal for any given K-ras mutation the ratio of mutant to normal DNA might then be expected to be relatively high. Kras is an oncogene, thus there is no reason to suppose that the normal copy of the gene should not be present in K-ras mutant tumour cells, unlike the occurrence of allele loss with tumour suppressor genes. Therefore, when taking into account the presence of normal DNA, the mutant sequences could actually be expected to account for only a small proportion of the total DNA of the sample and so go undetected by direct DNA sequencing. This is upheld by our observations and we therefore concluded that direct DNA sequencing was inappropriate for substantiating the ARMS results. This was confirmed by the second stage of validation where cloned amplicons of K-ras exon I were sequenced. Here, the DNA sequence data gave comparable results to those derived by ARMS. The results from this stage of validation therefore indicate the use of a method that will detect mutations that are under-represented against a background of wild-type alleles. The ARMS tests described herein were validated to a sensitivity of at least 1 mutant ras sequence in 103 wild-type sequences. In fact, control reactions for tumour DNA analysis had routinely lower ratios of mutant to wild-type input DNA. Other groups have reported tests based on the same principles as the ARMS tests described here. Detection levels of mutant ras sequences present at as low as 1 in 105 wildtype sequences have been reported. This ability of the technique to detect rare mutations in a background of normal DNA demonstrates its potential role in screening or in the monitoring of residual disease.
ARMS is a simple and accurate method and has several benefits over other PCRbased mutation detection systems. Specifically, the technique does not require the use of radioisotopes or the multiple probing of immobilised PCR amplicons or cloned PCR amplicons.
ARMS avoids the need for DNA sequencing of single-strand conformation polymorphism products, a procedure that could be expected to be constrained by sequence underrepresentation as discussed above. Similarly, under-represented mutant sequences could go undetected using PCR in conjunction with restriction fragment length polymorphism which is limited to low cycle numbers for the PCR to avoid false positive results. Studies that have examined the elimination of the BstNI restriction site at codon 12 in some K-ras mutations are severely limited because these tests rely on a BstNI restriction digest part way through PCR cycling. Previously generated amplicons therefore have the potential to cause carry-over contamination when PCR is resumed. ARMS can be performed under conditions in which carry-over contamination is avoided, as in the present study, allowing the use of high PCR cycle numbers and resulting in exceptionally high detection sensitivity.
One DNA sample analysed gave a positive result in more than one ARMS test.
Sample 1342 was derived from a Dukes' B rectal tumour from a 49 year old male. This sample gave a positive result with the K12 glycine (GGT) to arginine (CGT), serine (AGT) and alanine (GCT) tests. Direct sequence analysis was non-informative but DNA sequencing of each of the cloned PCR amplicons revealed a five nucleotide mutation. The normal sequence for codons 11 to 13 (GCT GGT GGC) was changed to GCC ACC AGC such that there is a reversed inversion of the last nucleotide of codon 11 to the first nucleotide of codon 13 resulting in a K12 glycine to threonine and K13 glycine to serine mutant protein. Such an occurrence would possibly be the result of an aberrant recombinogenic event. The DNA sequencing results therefore exclude the possibility that tumour 1342 is polyclonal for more than one K-ras point mutation. None of the tumours analysed were found to have more than one K-ras mutation, also the overall frequency of K-ras mutation does not increase significantly between Dukes' stages. This supports the model that K-ras mutation is a relatively early event in the progression of CRC through Dukes' stages A to C. Because only six adenomas were included in this study the exact timing of K-ras mutation in the adenoma to carcinoma progression was not addressed.
The male to female split of the total non-adenoma patients and the severity of their disease in association with their K-ras mutation status was examined. There was no evidence of differing proportions of tumours with or without K-ras mutations for the sexes either separately or overall in terms of the disease stage. There were more K-ras negative males compared to females, and the males presented with a more severe stage of the disease.
However, it should be realised that these observations could be influenced by other factors such as the catchment area from which the patients were drawn. Similarly, the difference in severity of the disease noted between the tumours of males and females could be related to the male/female attitude to the disease and to when they first consulted their primary care clinician.
The value of K-ras mutations as a marker of malignancy will depend on several factors, not least being the frequency of tumours of a given type, such as CRC, that carry the mutation. As this study has found the frequency of K-ras mutations to be approaching 40%, additional markers for CRC would be required for general screening purposes if all CRCs were to be identified using ARMS.
Tables Table 3: ARMS primer and common Drimer DNA sequences.
Primer Primer sequence (5' - 3') ARMS K12 Val TATCGTCAAGGCACTCTTGCCTACGCCTA ARMS K12 Cys CTGAATATAAACTTGTGGTAGTTGGAGCAT ARMS K12 Ser CTGAATATAAACTTGTGGTAGTTGGAGCCA ARMS K12 Arg CTGAATATAAACTTGTGGTAGTTGGAGCCC ARMS K12 Asp TATCGTCAAGGCACTCTTGCCTACGCCTT ARMS K12 Ala TATCGTCAAGGCACTCTTGCCTACGCCTG
Primer | Primer sequence (5' - 3') ARMS K13 Asp CGTGTATCGTCAAGGCACTCTT'GCCTACCT Common primer (cpA) GTACTGGTGGAGTATTTGATAGTGTATTAACC Common primer (cpB) CTCATGAAAATGGTCAGAGAAACCTTTATC Table 4: Cell lines used in the development and validation of the K-ras ARMS test.
Mutation Cell line Source 12Ser A549 ATCC ref CRL-7909 12Cys MIA PaCa-2 ATCC ref CRL-1420 12Arg PSN-1 Yamada H. et al., Biochem. Biophys. Res.
Commun., 140: 167-173, 1986 12Ala SW1116 ATCC ref CCL-233 12Val Capan 2 ATCC ref HTB-80 12Asp Panc 1 ATCC ref CRL-1469 13Asp HCTI 16 ATCC ref CCL-247 Table 5: Annealing temperature and PCR cvcle number for each K-ras mutation-specific ARMS primer.
Mutation Annealing Cycles temperature ( C) | 12Ser 60 36 12Cys 60 35 12Arg 61 35 12Ala 58 35 12Val 60 35 12Asp 63 35 13Asp 63 35 Table 6: The ARMS test. direct sequencing and clone analysis results grouped according to the ARMS primers found to generate K-ras amtlicons
Tumour Dukes' Stage ARMS result Direct DNA sequence result 6 C K12 Arg K12 Arg 23 C K12Cys wt 72 C K13 Asp K13 Asp 178 C K13 Asp K13 Asp 188 A K12 Val wt 202 B Asp wt 214 B K12 Cys wt 302 C K13 Asp wt 328 B K12 Asp K12 Asp 357 B K12 Asp K12 Asp 406 B K12 Val K12 Val 436 B K12Val wt 561 C K12 Val K12 Val 565 B K12 Asp wt 596 C K13 Asp wt 598 B K12 Asp K12 Asp 734 C K12 Asp K12Asp 777 B K12 Val wt 863 B K13 Asp K13 Asp 980 C K12 Ser K12 Ser 982 B K12 Ser K12 Ser 1076 C K12 Asp K12 Asp 1182 B K12 Val K12Val 1210 A K12 Val wt 1253 C K12 Cys wt 1257 A K12 Cys K12 Cys
Tumour Dukes' Stage ARMS result Direct DNA sequence result 1261 C K12 Val K12 Val 1271 C K12 Ser K12 Ser 1289 B K12 Val wt 1342 B K12 Ser+ Multiple Arg+Ala Table 7: Direct and cloned sequence results from tumour DNAs grouped according to the ARMS primer that initially characterized the K-ras mutation harbored bv the tumour.
Tumour ARMS result Direct sequence Frequency of Sequence of ARMS DNA result mutation in clones positive clones 1271 12 Ser 12 Ser 5 of 27 (19%) 12 Ser 1342 12 Ser unclear 16 of 28 (57%) 5 mutations 23 12 Cys wt 14 of 38 (37%) 12 Cys 214 12 Cys wt 12 of 33 (36%) 12 Cys 500 12 Cys wt 10 of 33 (30%) 12 Cys 530 12 Cys wt 2 of 34 (6%) 12 Cys 1253 12 Cys wt 21 of 43 (49%) 12 Cys 6 12 Arg 12 Arg 4 of 10 (40%) 12 Arg 957 wt wt 0 no +ve clones 1342 12 Arg unclear 0 no +ve clones 1342 12 Ala unclear not done not done 188 12 Val wt 8 of 39 (21%) 12 Val 436 12 Val wt 11 of 33 (33%) 12 Val 556 12 Val 12 Val 12 of 37 (32%) 12 Val 777 12 Val 12 Val 4 of 38 (10%) 12 Val 1210 12 Val wt 3 of 42 (7%) 12 Val 1289 12 Val 12 Val 10 of 38 (26%) 12 Val
Tumour ARMS result Direct sequence Frequency of Sequence of ARMS DNA result mutation in clones positive clones 177 12 Asp wt 15 of 41 (37%) 12 Asp 202 12 Asp wt 2 of 12 (21%) 12 Asp 357 12 Asp 12 Asp 20 of 41 (49%) 12 Asp 410 12 Asp wt 7 of 37 (19%) 12 Asp 546 12 Asp wt 3 of 39 (8%) 12 Asp 565 12 Asp wt 2 of 34 (6%) 12 Asp 302 13 Asp wt 4 of 41 (10%) 13 Asp 596 13 Asp wt 5 of 24 (21%) 13 Asp Table 8: Analysis of the frequencies of the K-ras mutations detected using ARMS from all samples from the CRC tumour bank.
Frequencies Frequencies Frequencies Tumour (male patients) (female patients) (all patients) K-ras+ve K-ras-ve K-ras+ve K-ras-ve K-ras+ve K-ras-ve Adenoma 1 of 4 3 of 4 0 of 2 2 of 2 1 of 6 5 of 6 (25.00 %) (75.00 %) (0.00 %) (100.00 % (16.66 %) (83.33 %) Dukes' A 5 of 15 10 of 15 8 of 16 8 of 16 13 of 31 18 of 31 (33.33 %) (66.66 %) (50.00 %) (50.00 %) (41.93 %) (58.06 %)| Dukes' B 24 of 74 50 of 74 23 of 61 38 of 61 47 of 135 88 of 135 (32.43 %) (67.57 %) (37.70 %) (62.30 %) (34.81 %) (65.19 %) Dukes'C 28 of 90 62 of 90 20 of 39 19 of 39 48 of 129 81 of 129 (31.11 %) (68.89 %) (51.28 %) (48.72 %) (37.21 %) (62.79 %)| Table 9: K13/K12 RAS Test.
Reagent mixes for each test are prepared and divided into aliquots (40 l) at 1.25 times final concentration.
Reagent Final concentration Upper control AAT primers 0.03125pom Lower control CF exon 4 primers 0.09375 pM ARMS primer 1.25pM Common primer 1.25pM Reagent Final concentration dNTPs 125pM ARMS buffer 1.25Xa alX ARMS buffer = 10mM Tris-HCl, (pH 8.3) 1.2mM MgCl2, 50mM KCl, 0.01% gelatin Table 10: K13/K12 RAS Test.
Reaction mix aliquots are transferred to reaction tubes according to individual test:
Mutation detected Tube type K12 VALINE Natural thin wall tubes K12 CYSTEINE Pink thin wall tubes K12 SERINE Green thin wall tubes K12 ARGININE Blue thin wall tubes K12 ASPARTATE Yellow thin wall tubes K12 ALANINE Purple thin wall tubes K13 ASPARTATE Orange thin wall tubes Table 11: Control primer sequences The lower control primers amplify part of exon four of the human cystic fibrosis transmembrane conductance regulator (CFTR) gene and the upper control primers amplify a part of each of exon II and intron m of the human al-antitrypsin gene.
Primer function Primer sequence (5' -3') Upper control 1 GGGCCTCAGTCCCAACATGGCTAAGAGGTG Upper control 2 CCCACCTTCCCCTCTCTCCAGGCAAATGGG Lower control 1 TATATGTGCCATGGGGCCTGTGCAAGGAAG Lower control 2 CTCCTACACCCAGCCATT'I'TTGGC Table 12: K13/K12 RAS test using "Amplitaq Gold" DNA polymerase To initiate each test AmpliTaq GoldTM (P. E. Applied Biosystems) is diluted to 5 l in 1X ARMS buffer and the test DNA (5 l) is added.
Mutation detected AmpliTaq GoldR (units/50 K12 VALINE 1.0 K12 CYSTEINE 1.0 K12 SERINE 0.5 K12 ARGININE 1.0 K12 ASPARTATE 0.6 K12 ALANINE 1.0 K13 ASPARTATE 0.6 Table 13: Final concentrations of the test reagents Thermal cycling conditions for all tests are: 94 C, 20 min; followed by 36 cycles of 94 C, 45 sec; 63 C, 45 sec; 72 C, 45 sec followed by a final incubation at 72 C, 10 nun.
Diagnostic and control amplicons are visualised after electrophoresis of reaction mix aliquots (20 l) through 3% Nusieve (FMC BioProducts) agarose gels against #X174/HaeIII digested DNA size marker.
Reagent Final concentration Upper control AAT primers 0.025pM Lower control CF exon 4 primers 0.07 5pM ARMS primer l.OpM Common primer 1.0 M Reagent Final concentration dNTPs 100pM ARMS buffer 1.125X Example 2 Identification of k12 ras mutations by arms linear extension ("ALEX"TM) Steps 1-15 Step 1: K-ras exon I is amplified by PCR in a final volume of 100 l. Two reactions are carried out for each sample, in each reaction one of the primers is biotinylated. This produces PCR products in which either the sense or anti-sense strand is biotinylated.
Table 14: Final concentrations of added reagents
Reagent Final concentration Forward primer 1.OOpM Reverse primer 1.00 M dNTPs 100pM ARMS buffer 1.125X Table 15: Primer sequences
Primer Primer sequence (5' - 3') Forward primer GTACTGGTGGAGTATTTGATAGTGTATAACC (Biotinylated)
Primer Primer sequence (5' - 3') Forward primer As above (Unmodified) Reverse primer CCTCTATTGTTGGATCATATTCGTCCA (Biotinylated) Reverse primer ACTCATGAAAATGGTCAGAGAAACCTTTATC (Unmodified) Step 2: Test DNA (1 l) is added.
Step 3: To initiate each test "AmpliTaq"TM DNA polymerase (P. E. Applied Biosystems) diluted to 2 units in 10 l in 2X ARMS buffer is added Step 4: Thermal cycling conditions for all tests are: 94 C, 5 min; prior to "Amplitaq"m DNA polymerase addition then 50 cycles of 94 C, 1 min; 60 C, 1 min; 72"C, 1 min followed by a final incubation at 720C, 10 min.
Step 5: Diagnostic amplicons are visualised after electrophoresis of reaction mix aliquots (5Cll) through 2% Nusieve 3:1 agarose (FMC BioProducts) agarose gels against #X174/HaeIII digested DNA size marker.
Step 6: The amplicon from the remainder of each reaction mixture is purified, for example, using QiaQuick PCR purification kit (Qiagen) according to the manufacturer's protocol.
Step 7: PCR products (about 1012 molecules) are immobilised in streptavidincoated microwell plates (Kodak) in Phosphate Buffered Saline at 370C for 60min.
Step 8: The immobilised products are denatured in 0.1M sodium hydroxide at ambient temperature for 15 min.
Step 9: The wells are washed with 6 x SSC to remove the non-biotinylated strand. (6 x SSC is prepared by appropriately diluting 20 x SSC; 20 x SSC = 3M sodium chloride, 0.3M sodium citrate, pH 7.0) Step 10: The appropriate ARMS primer (Table 13), (1 Rm) in 6 x SSC is hybridised to the immobilised single strand at 600C for 60min then the wells are washed with 6 x SSC.
Step 11: The ARMS primer is extended in 50F1 0.1 mM dNTPs, 5pM digoxigenin-l 1-2' deoxy-uridine-5'-triphosphate (Boehringer Mannheim) with 2 units AmpliTaq DNA polymerase (P. E. Applied Biosystems) in 1 x ARMS buffer at 60"C for 10 min.
Step 12: The extension reaction is terminated by the addition of 100Cll 0.5M EDTA and the wells are washed with 25mM Tris-HCl (pH 7.5), 125 mM NaCl, 0.3mM MgCl2 0.3% Tween-20 (TEST).
Step 13: Alkaline phosphatase conjugated anti- digoxigenin antibody (Boehringer Mannheim) is bound to extension products from the ARMS primers in TBST at 370C for 30 min. then wells are washed with 1M diethanolamine (pH 9.8).
Step 14: p-Nitrophenol phosphate (Concn lmg/ml) in 1M diethanolamine (pH 9.8) is added and incubated at ambient temperature for 30min.
Step 15: The optical density for each microplate microwell is measured at 405nm.
Example 3 Identification of k12 ras mutations by arms primer extension in combination with "Taqman'nM Table 16: Final concentrations of reagents Reagents are prepared according to the table in 45 l aqueous solution
Reagent Final concentration ARMS primer 1.0 M Common primer 1.0 M dNTPs 100 M ROX standardb Reagent Final concentration TaqMan probe 200nM MgCl2c 3.5mM Template DNA Approx 10 copies ARMS buffer 1.OXa alX ARMS buffer = 10mM Tris-HCl, (pH 8.3) 1.2mM MgCl2, 50mM KCl, 0.01% gelatin. bROX = 6-carboxyrhodamine. CIn addition to contribution from ARMS buffer.
Tables 17a and 17b: ARMS primer. common primer and "TaqMan"TM probe sequences Table 17a: Probe and Drimer sequences according to individual test
Probe/Primer Sequence (5' -3') "TaqMan"R probea CAAGAGTGCCTTGACGATACAGCTA Common primer (cpB) CTCATGAAAATGGTCAGAGAAACCTTTATC K12 CYSTEINE(ARMS) CTGAATATAAACTTGTGGTAGTTGGAGCAT K12 SERINE(ARMS) CTGAATATAAACTTGTGGTAGTTGGAGCCA K12 ARGININE(ARMS) CTGAATATAAACTTGTGGTAGTTGGAGCCC Table 17b: Probe and primer sequences according to individual test
Probe/Primer Sequence (5' - 3') "TaqMan"TM probea CTACCACAAGTTTATATTCAGTCATTTTCA Common primer (cpA) GTACTGGTGGAGTATTTGATAGTGTATTAACC K12 VALINE(ARMS) TATCGTCAAGGCACTCTTGCCTACGCCTA K12 ASPARTATE(ARMS) TATCGTCAAGGCACTCTTGCCTACGCCTT K12 ALANINE(ARMS) TATCGTCAAGGCACTCTTGCCTACGCCTG K13 ASPARTATE(ARMS) CGTGTATCGTCAAGGCACTCTTGCCTACCT aTaqManTM (Holland PM, et al., Proc Natl Acad Sci USA 1991; 88: 7276-7280.) probes were prepared by (P.E. Applied Biosystems) with FAM reporter dye (5') and TAMRA quencher dye (3'), [FAM = 6-carboxyfluorescein; TAMRA = N,N,N',N'-tetramethyl-6-carboxyrhodamine] "AmpliTaq Gold" (P. E. Applied Biosystems), 5 l (diluted to 400 units/ml in 1X ARMS buffer) is added.
Thermal cycling conditions for all tests are: 94 C, 20 min; followed by 50 cycles of 94 C, 41 sec; 60 C, 42 sec; 72 C, 52 sec.
Diagnostic fluorescence signals are measured using, for example, a 7700 Sequence Detector (P. E. Applied Biosystems).
Example 4 Identification of K-ras mutations by ARMS Linear Extension coupled with SERRS Steps 1-6.
Step 1: K-ra suitable capture sequences, known to persons skilled in the art, may be used. The capture probe is immobilised to a suitable support so that non-hybridised sequences remain free in solution and are easily removed. The capture sequence may be bound to the support by any convenient method. For example, the sequence may be biotinylated at its 5' end and bound to a support coated in avidin/streptavidin. Hybridisation conditions may employ any suitable combinations of time, temperature and buffer constituents, determined by consideration of the Tm of the probelK-ras target complex.
Capture of K-ras DNA may be achieved in a single hybridisation step. Alternatively, repeated rounds of hybridisation may be used; in this way the total amount of human genomic DNA hybridised is reduced but the relative level of K-ras to non- K-ras DNA bound after hybridisation, and removal of non-hybridised DNA, is greatly increased.
Step 2: Hybridised DNA may be eluted from the capture probe sequence using any convenient combination of temperature and/or buffer. Alternately if the capture probe sequence does not overlap with the site of prospective mutation analysis, mutation analysis by ARMS extension may be performed in situ i.e. directly on the immobilised K-ras sequence.
Step 3:The appropriate ARMS primer (Table 13), (1 un) in 6 x SSC is hybridised to the immobilised single strand at 60"C for 60min then the wells are washed with 6 x SSC.
Step 4:The ARMS primer is extended in 50p1 0.1 mM dNTPs with 2 units AmpliTaq DNA polymerase (P. E. Applied Biosystems) in 1 x ARMS buffer at 60"C for 10 nun.
Step 5:The extension reaction is terminated by the addition of 100Cl1 0.5M EDTA and the wells are washed with 25mM Tris-HCl (pH 7.5), 125 mM NaCl, 0.3mM MgCl2 0.3% Tween-20 (TBST).
Step 6: Extended K-ras primers may be detected by any suitable detection method such as surface enhanced ramon resonance spectroscopy (SERRS) whereby the extension product of an ARMS primer is hybridised to a complimentary sequence localised with a silver (AG) coated support. Hybridisation is detected by the change in SERRS signal measurable on hybridisation.
Example 5 System for the homogeneous detection of K-ras mutant sequences using ARMS coupled with amplicon detection by means of Taqman or Molecular Beacons.
SteDs 1-2.
Step 1: Reaction conditions are as described in Example 3. Amplicons may be detected using the "Taqman"TM probes shown in tables 17a and 17b. Alternatively, amplicons may be detected using the 2'-o-methyl-RNA Molecular Beacon probe sequences; 5' CGC GGU GCC UUG ACG AUA CAG CUA AUU CAG AAC GCG 3' and 5' CGC GGU GCU GAA AAU GAC UGA AUA UAA ACU UGU GGA COC G 3' at 100nM final test concentration.
Using the 2'-o-methyl-RNA Molecular Beacon described above the following data sets are derived: a) Real-time amplification plots are generated using templates comprised of either 103, 104 or 105 human diploid genome equivalents of DNA. Templates consist wholly of wild-type sequence or of a mixture of a single mutant species mixed with wild-type DNA in a ratio of 500 copies of wild-type to 1 copy of mutant. The mutant DNA is typically obtained from one of the cell lines described in Table 4. b) Reactions are replicated, by independent operators on different occasions, such that a final data set comprising up to 12 independent data points for each input DNA concentration and wild-type/mutant mix is obtained. c) An independent measurement of the concentration of DNA added is carried out in each case. This consists of a parallel real-time amplification of an amount (volume) of added DNA which is equal to that used in the ARMS reactions. The parallel amplification reaction acts as an independent verification of the initial DNA quantification carried out for the preparation of template mixes.
Step 2: The data sets generated in Step 1 are used to plot the control data graphs shown in Figure 5.
Example 6 Application of the real-time system for K-ras mutant sequence detection described in Example 5.
Step 1: Reaction mixes described in Example 3 containing either the "Taqman" > probe sequences described in Example 3 or the Molecular Beacon sequences described in Example 5 are prepared and dispensed into a 96 well plate as outlined in Figure 6.
Step 2: Spl volumes of purified DNA solution, derived from a source of clinical material, typically ranging from 103 to 105 total copy number/5p1 are added to each well of a single row in a 96 well plate as outlined in Figure 6. Amplification is carried out using appropriate conditions such as those described in Example 3 Step 3: Each row of a 96 well plate will produce a data set derived from a single DNA source. Interpretation of the data may conveniently be made with reference to the data sets, or similar data interpretations systems, described in Figure 5. Where initial DNA concentration falls below the desired range of 103 to 105 a preliminary amplification reaction of limited cycle number may be performed and the amplicons thus generated used in place of pure genomic DNA. Alternatively, where the initial DNA concentration is greater than 105 (estimable from the control reaction) an appropriate dilution from the neat sample may be performed followed by repeat analyses. In some circumstances it may desirable to perform a set of 8 reactions on two independent dilutions of the clinical sample derived DNA. This may be useful in situations where the sample contains impurities which may adversely affect the efficiency of the given ARMS reaction. However, since the control reaction itself comprises a mis-matched primer coupled with a common primer it is likely to be similarly affected and thus control for the presence of impurities.
Step 4: By analysing the results obtained, studies of the clinical relevance of the detection of K-ras mutations may be performed.
SEQUENCE LISTING (1) GENERAL INFORMATION: (i) APPLICANT: (A) NAME: Zeneca Limited (B) STREET: 15 Stanhope Gate (C) CITY: London (D) STATE: England (E) COUNTRY: United Kingdom (F) POSTAL CODE (ZIP): W1Y 6LN (G) TELEPHONE: 0171 3045000 (H) TELEFAX: 0171 3045151 (I) TELEX: 0171 3042042 (ii) TITLE OF INVENTION: Assay (iii) NUMBER OF SEQUENCES: 25 (iv) COMPUTER READABLE FORM: (A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS (D) SOFTWARE: PatentIn Release &num;1.0, Version &num;1.30 (EPO) (vi) PRIOR APPLICATION DATA: (A) APPLICATION NUMBER: GB 9715034.6 (B) FILING DATE: 18-JUL-1997 (2) INFORMATION FOR SEQ ID NO: 1: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 29 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1: TATCGTCAAG GCACTCTTGC CTACGCCTA 29 (2) INFORMATION FOR SEQ ID NO: 2: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 2: CTGAATATAA ACTTGTGGTA GTTGGAGCAT 30 (2) INFORMATION FOR SEQ ID NO: 3: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 3: CTGAATATAA ACTTGTGGTA GTTGGAGCCA 30 (2) INFORMATION FOR SEQ ID NO: 4: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 4: CTGAATATAA ACTTGTGGTA GTTGGAGCCC 30 (2) INFORMATION FOR SEQ ID NO: 5: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 29 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5: TATCGTCAAG GCACTCTTGC CTACGCCTT 29 (2) INFORMATION FOR SEQ ID NO: 6: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 29 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6: TATCGTCAAG GCACTCTTGC CTACGCCTG 29 (2) INFORMATION FOR SEQ ID NO: 7: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 7: CGTGTATCGT CAAGGCACTC TTGCCTACCT 30 (2) INFORMATION FOR SEQ ID NO: 8: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 32 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8: GTACTGGTGG AGTATTTGAT AGTGTATTAA CC 32 (2) 1NFORMATION FOR SEQ ID NO: 9: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9: CTCATGAAAA TGGTCAGAGA AACCTTTATC 30 (2) INFORMATION FOR SEQ ID NO: 10: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10: GGGCCTCAGT CCCAACATGG CTAAGAGGTG 30 (2) INFORMATION FOR SEQ ID NO: 11: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11: CCCACCTTCC CCTCTCTCCA GGCAAATGGG 30 (2) INFORMATION FOR SEQ ID NO: 12: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12: TATATGTGCC ATGGGGCCTG TGCAAGGAAG 30 (2) INFORMATION FOR SEQ ID NO: 13: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 24 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 13: CTCCTACACC CAGCCATTT TGGC 24 (2) INFORMATION FOR SEQ ID NO: 14: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 41 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14: CTGGATCTAG ACTCATGAAA ATGGTCAGAG AAACCTTTAT C 41 (2) INFORMATION FOR SEQ ID NO: 15: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 43 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15: CCTCGGAATT CGTACTGGTG GAGTA?TTGA TAGTGTATTA ACC 43 (2) INFORMATION FOR SEQ ID NO: 16: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 17 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 16: GTTTTCCCAG TCACGAC 17 (2) INFORMATION FOR SEQ ID NO: 17: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 17 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 17: CAGGAAACAG CTATGAC 17 (2) INFORMATION FOR SEQ ID NO: 18: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 27 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 18: CCTCTATTGT TGGATCATAT TCGTCCA 27 (2) INFORMATION FOR SEQ ID NO: 19: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 31 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19: ACTCATGAAA ATGGTCAGAG AAACCTTTAT C 31 (2) INFORMATION FOR SEQ ID NO: 20: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 25 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 20: CAAGAGTGCC TTGACGATAC AGCTA 25 (2) INFORMATION FOR SEQ ID NO: 21: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 30 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 21: CTACCACAAG TT'TATATTCA GTCATTITCA 30 (2) INFORMATION FOR SEQ ID NO: 22: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 24 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 22: TGAAAATGGT CAGAGAAACC TTTA 24 (2) INFORMATION FOR SEQ ID NO: 23: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 36 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 23: CGCGGUGCCU UGACGAUACA GCUAAUUCAG AACGCG 36 (2) INFORMATION FOR SEQ ID NO: 24: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 40 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 24: CGCGGUGCUG AAAAUGACUG AAUAUAAACU UGUGGACGCG 40 (2) INFORMATION FOR SEQ ID NO: 25: (i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 18 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: other nucleic acid (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 25: GGAGCTGGTG GCGTAGGC 18

Claims (9)

  1. CLAIMS 1. A diagnostic method for the detection of K-ras mutations in cancer, which method comprises contacting a test sample of nucleic acid with a diagnostic primer for a K-ras mutation in the presence of appropriate nucleotide triphosphates and an agent for polymerisation, such that the diagnostic primer is extended only when a K-ras mutation is present in the sample; and detecting the presence or absence of a diagnostic primer extension product.
  2. 2. A diagnostic method as claimed in claim 1 wherein the K-ras mutations detected comprise one or more of the following mutations: K-ras codon 12 GGCoAGT (glycine # serine) K-ras codon 12 GGToCGT (glycine # arginine) K-ras codon 12 GGToTGT (glycine -4 cysteine) K-ras codon 12 GGTeGAT (glycine o aspartic acid) K-ras codon 12 GGTeGCT (glycine < alanine) K-ras codon 12 GGT#GTT (glycine -4 valine) K-ras codon 13 GGCoGAC (glycine -4 aspartic acid)
  3. 3. A diagnostic method as claimed in claim 1 and claim 2 wherein the diagnostic primer for a K-ras mutation is used with a further amplification primer in one or more cycles of PCR amplification.
  4. 4. A diagnostic method as claimed in any one of claims 1-3 wherein the test sample of nucleic acid is firstly amplified.
  5. 5. A diagnostic method as claimed in claim 1 wherein the diagnostic primer extension product is detected using a non-isotopic labelling method or real-time detection.
  6. 6. A diagnostic method as claimed in claim 1 wherein two or more diagnostic primers are used in the same reaction vessel.
  7. 7. Diagnostic primers for the detection of K-ras mutations comprising the sequences given below which detect the following mutations respectively: CTGAATATAAACTTGTGGTAGTTGGAGCCA K12 Serine CTGAATATAAACTTGTGGTAGTTGGAGCCC K12 Arginine CTGAATATAAACTTGTGGTAGTTGGAGCAT K 12 Cysteine TATCGTCAAGGCACTCTTGCCTACGCCTT K12 Aspartic acid TATCGTCAAGGCACTCTTGCCTACGCCTG K12 Alanine TATCGTCAAGGCACTCTTGCCTACGCCTA K12 Valine CGTGTATCGTCAAGGCACTCTTGCCTACCT K13 Aspartic acid
  8. 8. A diagnostic kit comprising one or more of the diagnostic primers as claimed in claim 7, nucleotide triphosphates, polymerase, buffer solution and instructions for use.
  9. 9. A diagnostic kit as claimed in claim 8 comprising all seven diagnostic primers.
GB9815224A 1997-07-18 1998-07-15 Diagnostic assay for cancer Expired - Fee Related GB2327497B (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
GB9715034A GB9715034D0 (en) 1997-07-18 1997-07-18 Assay

Publications (3)

Publication Number Publication Date
GB9815224D0 GB9815224D0 (en) 1998-09-09
GB2327497A true GB2327497A (en) 1999-01-27
GB2327497B GB2327497B (en) 1999-12-08

Family

ID=10815987

Family Applications (2)

Application Number Title Priority Date Filing Date
GB9715034A Pending GB9715034D0 (en) 1997-07-18 1997-07-18 Assay
GB9815224A Expired - Fee Related GB2327497B (en) 1997-07-18 1998-07-15 Diagnostic assay for cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
GB9715034A Pending GB9715034D0 (en) 1997-07-18 1997-07-18 Assay

Country Status (2)

Country Link
GB (2) GB9715034D0 (en)
WO (1) WO1999004037A1 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000066783A2 (en) * 1999-05-04 2000-11-09 Ortho-Clinical Diagnostics, Inc. Rapid and efficient capture of dna from sample without using cell lysing reagent
WO2001042781A3 (en) * 1999-12-07 2002-05-10 Exact Sciences Corp Supracolonic aerodigestive neoplasm detection
US6586177B1 (en) 1999-09-08 2003-07-01 Exact Sciences Corporation Methods for disease detection
WO2004029288A2 (en) * 2002-09-26 2004-04-08 Novartis Ag Pcr-based diagnostic method of detecting a mutation in the b-raf gene
US7262006B1 (en) 2000-05-01 2007-08-28 Ortho-Clinical Diagnostics, Inc. Rapid and efficient capture of DNA from sample without using cell lysing reagent
WO2010071821A1 (en) * 2008-12-19 2010-06-24 Abbott Laboratories Diagnostic test for mutations in codons 12-13 of human k-ras
US7776524B2 (en) 2002-02-15 2010-08-17 Genzyme Corporation Methods for analysis of molecular events
US7833757B2 (en) * 2003-09-23 2010-11-16 Universitael Posidam Method for conducting non-invasive early detection of colon cancer and/or of colon cancer precursor cells
US7897356B2 (en) 2008-11-12 2011-03-01 Caris Life Sciences Methods and systems of using exosomes for determining phenotypes
US8700335B2 (en) 2006-05-18 2014-04-15 Caris Mpi, Inc. System and method for determining individualized medical intervention for a disease state
US8768629B2 (en) 2009-02-11 2014-07-01 Caris Mpi, Inc. Molecular profiling of tumors
US9128101B2 (en) 2010-03-01 2015-09-08 Caris Life Sciences Switzerland Holdings Gmbh Biomarkers for theranostics
US9469876B2 (en) 2010-04-06 2016-10-18 Caris Life Sciences Switzerland Holdings Gmbh Circulating biomarkers for metastatic prostate cancer
JP2017108735A (en) * 2015-12-11 2017-06-22 東洋紡株式会社 Reagent for nucleic acid amplification and nucleic acid amplification method

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2424584A1 (en) * 2000-10-02 2002-04-11 Ortho-Clinical Diagnostics, Inc. Detection of ras mutations
NZ602920A (en) * 2010-04-12 2014-07-25 Response Genetics Inc Kras primers and probes
EP2607495A1 (en) 2011-12-23 2013-06-26 Genomica S.A.U. Method for detection of KRAS mutations

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991013075A2 (en) * 1990-02-16 1991-09-05 Orion-Yhtymä Oy Method and reagent for determining specific nucleotide variations
US5137806A (en) * 1989-12-11 1992-08-11 Board Of Regents, The University Of Texas System Methods and compositions for the detection of sequences in selected DNA molecules
GB2299166A (en) * 1993-12-16 1996-09-25 Maurice Stroun Method for diagnosing cancer
WO1996040995A1 (en) * 1995-06-07 1996-12-19 Trustees Of Dartmouth College Detection of gene sequences in biological fluids
WO1997019191A1 (en) * 1995-11-17 1997-05-29 Oegreid Dagfinn Method for detection of ki-ras mutations and kit to perform it

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2134552A1 (en) * 1992-04-27 1993-11-11 George D. Sorenson Detection of gene sequences in biological fluids

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5137806A (en) * 1989-12-11 1992-08-11 Board Of Regents, The University Of Texas System Methods and compositions for the detection of sequences in selected DNA molecules
WO1991013075A2 (en) * 1990-02-16 1991-09-05 Orion-Yhtymä Oy Method and reagent for determining specific nucleotide variations
GB2299166A (en) * 1993-12-16 1996-09-25 Maurice Stroun Method for diagnosing cancer
WO1996040995A1 (en) * 1995-06-07 1996-12-19 Trustees Of Dartmouth College Detection of gene sequences in biological fluids
WO1997019191A1 (en) * 1995-11-17 1997-05-29 Oegreid Dagfinn Method for detection of ki-ras mutations and kit to perform it

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Oncogene (1991) 6 857-862 (Stork P. et al.) *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000066783A3 (en) * 1999-05-04 2002-01-03 Ortho Clinical Diagnostics Inc Rapid and efficient capture of dna from sample without using cell lysing reagent
US7615346B2 (en) 1999-05-04 2009-11-10 Ortho-Clinical Diagnostics, Inc. Rapid and efficient capture of DNA from sample without using cell lysing reagent
WO2000066783A2 (en) * 1999-05-04 2000-11-09 Ortho-Clinical Diagnostics, Inc. Rapid and efficient capture of dna from sample without using cell lysing reagent
US6586177B1 (en) 1999-09-08 2003-07-01 Exact Sciences Corporation Methods for disease detection
US7981612B2 (en) 1999-12-07 2011-07-19 Mayo Foundation For Medical Education And Research Methods of screening for supracolonic neoplasms based on stool samples containing a nucleic acid marker indicative of a neoplasm
WO2001042781A3 (en) * 1999-12-07 2002-05-10 Exact Sciences Corp Supracolonic aerodigestive neoplasm detection
US7262006B1 (en) 2000-05-01 2007-08-28 Ortho-Clinical Diagnostics, Inc. Rapid and efficient capture of DNA from sample without using cell lysing reagent
US8409829B2 (en) 2002-02-15 2013-04-02 Esoterix Genetic Laboratories, Llc Methods for analysis of molecular events
US7776524B2 (en) 2002-02-15 2010-08-17 Genzyme Corporation Methods for analysis of molecular events
WO2004029288A2 (en) * 2002-09-26 2004-04-08 Novartis Ag Pcr-based diagnostic method of detecting a mutation in the b-raf gene
WO2004029288A3 (en) * 2002-09-26 2004-09-02 Novartis Ag Pcr-based diagnostic method of detecting a mutation in the b-raf gene
US7833757B2 (en) * 2003-09-23 2010-11-16 Universitael Posidam Method for conducting non-invasive early detection of colon cancer and/or of colon cancer precursor cells
US8700335B2 (en) 2006-05-18 2014-04-15 Caris Mpi, Inc. System and method for determining individualized medical intervention for a disease state
US7897356B2 (en) 2008-11-12 2011-03-01 Caris Life Sciences Methods and systems of using exosomes for determining phenotypes
WO2010071821A1 (en) * 2008-12-19 2010-06-24 Abbott Laboratories Diagnostic test for mutations in codons 12-13 of human k-ras
US8768629B2 (en) 2009-02-11 2014-07-01 Caris Mpi, Inc. Molecular profiling of tumors
US9128101B2 (en) 2010-03-01 2015-09-08 Caris Life Sciences Switzerland Holdings Gmbh Biomarkers for theranostics
US9469876B2 (en) 2010-04-06 2016-10-18 Caris Life Sciences Switzerland Holdings Gmbh Circulating biomarkers for metastatic prostate cancer
JP2017108735A (en) * 2015-12-11 2017-06-22 東洋紡株式会社 Reagent for nucleic acid amplification and nucleic acid amplification method
JP2021141898A (en) * 2015-12-11 2021-09-24 東洋紡株式会社 Reagent for nucleic acid amplification, and nucleic acid amplification method

Also Published As

Publication number Publication date
GB9715034D0 (en) 1997-09-24
GB2327497B (en) 1999-12-08
GB9815224D0 (en) 1998-09-09
WO1999004037A1 (en) 1999-01-28

Similar Documents

Publication Publication Date Title
US5871925A (en) Tumor classification as indication of hereditary non-polyposis colorectal carcinoma
GB2327497A (en) Diagnostic assay for cancer
US6479234B1 (en) Detection of hypermutable nucleic acid sequence in tissue and body fluids
Goessl et al. DNA alterations in body fluids as molecular tumor markers for urological malignancies
JP4157164B2 (en) Changes in genes associated with prostate cancer
US6291163B1 (en) Method for detecting cell proliferative disorders
US20140206855A1 (en) Methods and compositions for detecting cancers associated with methylation of hmlh1 promoter dna
US20090181400A1 (en) Method of Detection of Prostate Cancer
EP0954608A1 (en) Methylation specific detection
US20080213791A1 (en) Materials and methods for assaying for methylation of CpG islands associated with genes in the evaluation of cancer
CA2519456A1 (en) Aberrantly methylated genes in pancreatic cancer
US20130323736A1 (en) Polynucleotide primers
US6001569A (en) Methods of screening for Crohn&#39;s disease using TNF microsatellite alleles
TW202102689A (en) Multi-gene combined detection reagent
CA3146980A1 (en) Genetic marker combination and use thereof
WO1996017080A1 (en) Detecting tumours
CA2067114A1 (en) Sensitive method for measurement of chimeric transcripts of dna containing translocations
US20040106109A1 (en) Detection of ras mutations
US6255049B1 (en) Detection of metastatic cancer cells using PCTA-1
US20030104454A1 (en) Method for detection of DNA methyltransferase RNA in plasma and serum
WO2001053535A2 (en) Method of detecting neoplastic, hyperplastic, cytologically dysplastic and/or premalignant cellular growth or proliferation
US7611841B2 (en) Method for detecting methylation of promoter using restriction enzyme and DNA chip
JP2006510870A (en) Molecular diagnosis and prognosis of carcinoma
JPWO2021004214A5 (en)
WO2002029005A2 (en) Detection of ras mutations

Legal Events

Date Code Title Description
732E Amendments to the register in respect of changes of name or changes affecting rights (sect. 32/1977)
PCNP Patent ceased through non-payment of renewal fee

Effective date: 20020715