CA2295333C - Assaying nucleotides in solution using a fluorescent intensity quenching effect - Google Patents

Assaying nucleotides in solution using a fluorescent intensity quenching effect Download PDF

Info

Publication number
CA2295333C
CA2295333C CA002295333A CA2295333A CA2295333C CA 2295333 C CA2295333 C CA 2295333C CA 002295333 A CA002295333 A CA 002295333A CA 2295333 A CA2295333 A CA 2295333A CA 2295333 C CA2295333 C CA 2295333C
Authority
CA
Canada
Prior art keywords
nucleotide sequence
detecting
probe
sequence according
probes
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.)
Expired - Fee Related
Application number
CA002295333A
Other languages
French (fr)
Other versions
CA2295333A1 (en
Inventor
Yuan Min Wu
Eileen Xiao-Feng Nie
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.)
Ingeneus Corp
Original Assignee
Ingeneus Corp
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 Ingeneus Corp filed Critical Ingeneus Corp
Publication of CA2295333A1 publication Critical patent/CA2295333A1/en
Application granted granted Critical
Publication of CA2295333C publication Critical patent/CA2295333C/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
    • C12Q1/6813Hybridisation assays
    • C12Q1/6832Enhancement of hybridisation reaction
    • 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
    • C12Q2600/156Polymorphic or mutational markers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S435/00Chemistry: molecular biology and microbiology
    • Y10S435/81Packaged device or kit

Abstract

The invention provides a method for rapidly, economically and efficently sequencing and assaying nucleotides in a liquid medium using laser induced fluorescence of antisense probes, including PNA probes. Fluorescent intensity of the resulting medium is inversely proportional to the hybridization efficiency of the probes with respect to t he target sequence. The method is particularly advantageous in not requiring separation of unhybridized probes and hybridization complexes prio r to detection. The method can be used to identify accessible regions in folded nucleotide sequences, to determine the number of mismatche d pairs in a hybridization complex, and to map genomes.

Description

ASSAYING NUCLEOTIDES IN SOLUTION USING
A FLUORESCENT INTENSITY QUENCHING EFFECT
SPECIFICATION
BACKGROUND OF THE INVENTION
1. Field of Invention The invention relates to methods of sequencing or assaying nucleotides in solution, without the need for solid support or adjacent double stranded nucleotide construct, using a fluorescent intensity quenching effect exhibited by antisense probes, such as peptide nucleic acid (PNA) probes.
2. Description of Related Art PNAs are polyamide analogs of DNA and RNA. See, e.g., U.S. Patent No. 5,539,082 to Nielsen et al. Nielsen et al.
discloses that PNAs mimic natural polynucleotides by binding complementary single stranded (ss) DNA and RNA strands. PNAs generally comprise ligands linked to a peptide backbone.
Representative ligands include either the four main naturally occurring DNA bases (i.e., thymine, cytosine, adenine or guanine) or other naturally occurring nucleobases (e. g., inosine, uracil, 5-methylcytosine or thiouracil) or artificial bases (e. g., bromothymine, azaadenines or azaguanines, etc.) attached to a peptide backbone through a suitable linker.
Probes comprising PNA sequences have been employed to detect target nucleotide sequences. U.S. Patent No. 5,503,980 to Cantor suggests employing PNA probes in a method of sequencing a nucleic acid by hybridizing the nucleic acid with a set of PNA probes containing random, but determinable, base sequences within the single stranded portion adjacent to a double stranded portion, wherein the single stranded portion of the set preferably comprises every possible combination of sequences over a predetermined range.
Hybridization occurs by complementary recognition of the single stranded portion of a target with the single stranded portion of the probe and is thermodynamically favored by the presence of adjacent double strandedness of the probe.
However, although Cantor discloses that the nucleic acids can be PNAs, it does not disclose or suggest utilizing such probes in the absence of a solid support. Moreover, the CONFIRMATION COPY

present invention does not require the adjacent construct of DNA material being tested.
In addition to teaching the use of a solid support like Cantor, Perry-O'Keefe et al., "Peptide Nucleic Acid Pre-Gel Hybridization: An Alternative to Southern Hybridization,"
93 Proc. Natl. Acad. Sci. USA 14670 (December 1996) also teaches that PNA does not generally bind well to double stranded DNA (dsDNA) . See Perry-O'Keefe et al. at page 14673, footnote. Moreover, the homopyrimidine PNA constructs which have been found to bind dsDNA well would not be useful as probes. Applicants have discovered that the qualification which suggests that only homopyrimidine can bind with dsDNA
by strand invasion is incorrect and arises from the hybridiza-tion conditions employed.
Smulevitch et al., "Enhancement of Strand Inversion by Oligonucleotides Through Manipulation of Backbone Charge,"
14 Nature Biotechnology 1700 (Dec. 1996) (disclosed in Landsdorp, "Close Encounters of the PNA Kind," 14 Nature Biotechnology 1653 (Dec. 1996)) discloses using PNA primers to hybridize with dsDNA. However, Smulevitch et al. teaches the use of gels in detecting hybridization, and does not suggest the use of fluorescent markers.
Many types of sample analysis rely upon the fluorescent properties of a marker. Fluorescence occurs when a molecule excited by light of one wavelength returns to the unexcited (ground) state by emitting light of a longer wavelength. The exciting and emitted light, being of different wavelengths, can be separated from one another using optical filters, a camera or a CCD. Fluorescence has been used to visualize certain molecules (and hence structures) by light microscopy for many years, and is also used in other analytical techniques, such as flow cytometry. Further, the emission of fluorescence showing different colors can be detected by a human eye, a camera, a charge coupled device (CCD) or a photomultiplier.
For example, U.S. Patent No. 5,594,138 to Dykstra et al. discloses a method of fluorescent detection of a nucleic acid. The method comprises contacting the nucleic acid with a fluorescent marker that is a bis-dicationic aryl furan compound and exposing the nucleic acid to light at a frequency inducing fluorescence of the fluorescent marker.
The fluorescent marker may be conjugated to a nucleotide sequence as a probe for hybridization studies, or it may be conjugated to numerous reagents for in situ labeling studies.
Hybridization occurs on a solid support.
U.S. Patent No. 4,963,477 to Tchen discloses a probe of high sensitivity containing a modified nucleic acid, which can be recognized by specific antibodies.
Fluorescent In Situ Hybridization (FISH) is a technique comprising detecting fluorescent probe binding to human chromosomes by attaching DNA to a solid support, such as a glass slide. See, e.g., K.H. Andy Choo, Ed., '~In Situ Hybridization Protocols," Chapters 2 and 4 (Humana Press, Totowa, NJ, 1994). Like all other conventional detection methods comprising hybridization with probes, this method relies on the solid support to keep the two complementary strands of DNA apart while the probe hybridizes with one of the strands. In addition, FISH requires a complicated buffer and temperature control protocol, with overnight incubation.
U.S. Patents Nos. 5,538,848 to Livak et al. and 9,220,450 to Maggio disclose fluorescence-based detection of nucleotide sequences using oligonucleotide probes in solution; however, these patents require the use of a quenching agent in combination with a reporting agent, so as to distinguish between the signals generated by hybridized probes and unhybridized probes. Livak et al. also requires the use of enzymes in its disclosed method. Quenching agents and enzymes add complexity and expense to the methods.
WO 94/12665 pertains to fluorescent anisotropy techniques for detecting nucleotide hybridization. See, e.g., page 5, lines 1-8; page 19, lines 5-9; and page 24, lines 7-12 of the reference. The inverse of fluorescent AMENDED SHEET
IPEA/EP

,CA 02295333 1999-12-29 .. .
_ :._ ....
::
anisotropy or polarization measurements is not proportional to a number of base mismatches. That is, fluorescent anisotropy or polarization measurements increase with increasing hybridization.
EP 599,337 discloses several techniques for detecting nucleotide hybridization, including fluorescent polarization (see, e.g., page 4, lines 11-18), which is not relevant to the instantly claimed invention for the reasons discussed above.
In addition, EP 599,337 claims and uiscloses methods in which a signal is detected based on charge-transfer interaction between adjacent "reagents" added to the hybridization solution or attached to the probe and/or target, unlike the present invention. Moreover, EP 599,337 fails to disclose or suggest a quenching effect that is a predictable function of the number of base mismatches over a selected range, as in the present invention.
Cooper et al., 29 Biochemistry 9261-68 (1990), like EP
599,337, discloses charge-transfer techniques, and thus, teaches away from the instant invention for at least the same reasons. Cooper et al. shows a single example which incidentally shows that fluorescent intensity is reduced at about 525 nm upon hybridization of a fluorescein labeled strand to its unlabeled complement, but this does not suggest a fluorescent intensity quenching effect that is a predictable function of the number of base mismatches over a selected range.
U.S. Patent No. 5,332,659 to Kidwell discloses a method for detecting nucleotide sequences in solution using probes comprising at least two fluorophore moieties. The fluorophores must be selected to electronically interact with each other when close enough to vary the wavelength dependence of their spectra. Unhybridized probes are much more flexible than probes hybridized to the target sequence, and consequently the two fluorophore moieties on each probe AMENDED SHEET
IPEA/EP

are more likely to be close to each other when the probe is unhybridized than when the probe is hybridized. Thus, a change in emission wavelength correlated with free probe can be monitored as an indication of the amount of free probe in the sample.
Until the present invention, however, it has not been possible to rapidly test for the presence of nucleotide sequences in solution using a method which does not destroy the sample, is less hazardous to laboratory personnel than radiation based assays, does not require the cost and delay of separating unhybridized probes from hybridization complexes, does not require the provision of quenching agents, does not require the provision of enzymes, does not require the provision of multiple interactive reporting moieties on each probe and is readily automated. Time and cost efficient detection of mutant genetic sequences has been the rate limiting step in correlating mutant genotypes with altered phenotypes. Although conventional DNA sequencing methods have been considered to be the most accurate means of identifying mutations, these methods have been relatively slow and labor intensive, and are not particularly well-suited to rapidly screening large numbers of samples of genomic DNA for purposes including medical diagnosis, genomic sequencing and mapping.
SUMMARY OF THE INVENTION
The present invention provides methods for detecting nucleic acid sequences and/or determining sequence information, including genomic sequence information, from nucleic acids using probes comprising DNA, RNA, DNA analogs and/or RNA analogs (hereinafter ~~antisense probes").
The invention provides a method for detecting at least one single stranded or double stranded nucleotide sequence in a liquid medium. The method comprises: (1) adding to the liquid medium at least one antisense probe having at least AMENDED SHEET
IPEA/EP ~

one marker to form at least one hybridization complex with at least one nucleotide sequence in the medium; and (2) detecting the at least one nucleotide sequence by detecting at least one signal that is inversely proportional to an amount 5 of the at least one hybridization complex in the liquid medium and proportional to an amount of the at least one probe unhybridized in the liquid medium. The method can be conducted without separating unhybridized probes from the at least one hybridization complex prior to signal detecting, without providing a signal quenching agent on the at least one probe or on the at least one nucleotide sequence, and without the use of enzymes. The method can be employed to map genomes by screening a genomic library with a plurality of probes.
BRIEF DESCRIPTION OF THE DRAWINGS
The invention will be described in conjunction with the following drawings in which like reference numerals designate like elements and wherein:
Figure lA is a schematic depiction of an embodiment of an apparatus according to the invention;
Figures iB, 1C, 2, 3, and 4 are fluorescent spectra;
Figure 5 is a schematic depiction of an embodiment of an apparatus according to the invention; and Figures 6, 7A, 7B, 8A and 8B are fluorescent spectra.
DETAILED DESCRIPTION OF PREFERRED EMBODIMENTS
The invention utilizes antisense probes to detect and/or characterize nucleotide sequences in a sample. The expression "antisense probes" as used herein includes any probe capable of specifically binding to a nucleotide sequence having a base sequence complementary to the base sequence of the probe.
The antisense probes of the invention can be complementary to either strand of dsDNA, for example. PNA probes are most preferred, and are the focus of much this specification, but the invention is not limited thereto.
PNA probes are able to recognize dsDNA by hybridizing one strand, thereby presumably binding with the other strand to generate a PNA-DNA complex. Such recognition can take place to dsDNA target sequences 20 or more base pairs long.
Probe sequences having any length from 8 to 20 bases are preferred since this is the range within which the smallest _ unique DNA sequences of prokaryotes and eukaryotes are found.
Probes of 12 to 18 bases are particularly preferred since this is the length of the smallest unique sequences in the human genome. However, a plurality of shorter probes can be used to detect a nucleotide sequence having a plurality of non-unique target sequences therein, which combine to uniquely identify the nucleotide sequence.
At least the PNA probes of the invention are able to form triplex complexes with dsDNA and duplex complexes with RNA or ssDNA. The PNA probes of the invention are also able to form triplex complexes wherein a first PNA probe binds with RNA or ssDNA and a second ssDNA strand binds with the resulting duplex complex. See, e.g., Egholm et al., "PNA
Hybridizes to Complementary Oligonucleotides Obeying the Watson-Crick Hydrogen-Bonding Rules," 365. Nature 566 (1993), and Tomac et al., "Ionic Effects on the Stability and Conformation of Peptide Nucleic Acid Complexes," 118 J.Am.Chem.Soc. 5544 (1996).
In the PNA probes according to the invention, the bases attached to the polyamide backbone are primarily naturally occurring nucleobases attached at the position required by probe manufacture. Alternatively, the bases may be non-naturally occurring nucleobases (nucleobase analogs), other base-binding moieties, aromatic moieties, (C1-C4) alkanoyls, hydroxyls or even hydrogens. It will be understood that the term nucleobase includes nucleobases bearing removable protecting groups. Furthermore, at least one base on the polyamide skeleton can be replaced with, or substituted with, a DNA intercalator, a reporter ligand such as, for example, a fluorophore, radio label, spin label, hapten, or a protein-recognizing ligand such as biotin. Preferred detectable labels include a radioisotope, a stable isotope, an enzyme, a fluorescent chemical, a luminescent chemical, a chromatic chemical, a metal, an electric charge, or a spatial structure.
In particularly preferred embodiments, the probe comprises an antisense sequence covalently bonded to a fluorescent marker, which fluoresces when irradiated with a laser.
Preferred fluorescent markers include biotin, rhodamine and fluorescein.
The intensity of the fluorescence is proportional to the amount of free probe in solution and inversely proportional to the amount of hybridized probe in solution. That is, there is a quenching effect associated with hybridization of the probe and target sequence. The quenching effect varies with the marker selected. This effect enables the method of the invention to detect hybridization without employing a quenching agent on the probe (to quench unhybridized probe signal) or on the target sequence (to quench hybridized probe signal), as required by, e.g., Livak et al. and Maggio, supra.
Unlike Kidwell, supra, the instant invention does not require a plurality of electronically interacting fluorophores on each probe, because the fluorescent intensity quenching effect detected by the instant invention is not the same as the emission wavelength shift detected in Kidwell, which is caused by intramolecular excimer formation between adjacent fluorophores. The quenching effect of the instant invention is apparent with only one fluorophore per probe (although a plurality of fluorophores per probe are contemplated for certain embodiments).
In certain embodiments, the fluorescent marker is provided at the 5' terminal of the probe with a short linker to minimize interaction with the probe. However, the position of the marker within the probe does not appear to be particularly significant.
In order to distinguish a mutant nucleotide sequence from a reference nucleotide sequence, wherein the two sequences differ by as little as a single base, it is preferred to design the probe so that the mutant portion of the mutant nucleotide corresponds to the center of the probe. This design results in a higher hybridization yield and a more stable hybrid than when the mutant portion of the nucleotide corresponds to a terminus of the probe, since the bonding mismatch between probe and nucleotide is located centrally within the probe.
Probes are added to a liquid medium suspected of containing at least one nucleotide sequence, and/or a mutant version of the at least one sequence. The liquid medium can be any conventional medium known to be suitable for preserving nudleotides. See, e.g., Sambrook et al., "Molecular Cloning:
A Lab Manual," 2d (1989). For example, the liquid medium can comprise nucleotides, water, buffers and surfactants.
l0 The nucleotides in the liquid medium can be obtained from clinical samples by any conventional method, including an automated method. Examples of such methods are summarized in, e.g., Sambraok et al., Vol. 2, pp. 9.16-9.19 and 7.6 to 7.7. An example of an automated nucleic acid purifying apparatus is the BioRobot* 9600 manufactured by Quiagen (Chatsworth, CA, USA).
For example, a variety of diseases are known to be linked with the presence of. mutant DNA in an individual's genome.
If the sequences of the wild type DNA and the mutant DNA are known, it is possible to isolate these nucleotide sequences from clinical samples using conventional technology. PCR
is the preferred method of amplifying nucleotides from clinical samples. PCR is conducted using a primer which is capable of amplifying the wild type DNA and the mutant DNA.
The nucleotide sequences are added to the liquid medium in a known concentration, since the concentration can affect the magnitude of the signal (e. g., fluorescent intensity) generated in subsequent steps in the inventive method. The nucleotide concentration can be determined by; e.g., measuring the UV absorption at 260 nm.
The isolated nucleotides are added to the liquid medium and denatured prior to being detected. Preferably, the denaturation is conducted at about 90°C to about 1o0°C from about:30 seconds to about 5 hours in the presence of PNA probe.
Preferably, probes are added to the liquid medium in a concentration 0.05 to 100 times the concentration of the nucleotide sequence to be detected.
Trademark*
Hybridization between complementary bases occurs under a wide variety of conditions having variations in temperature, salt concentration, electrostatic strength, and buffer composition. Examples of these conditions and methods for applying them are known in the art. See, e.g. , Perry-O'Keefe et al., Egholm et al., Tomac et al., Sambrook et al., Vol.
2 pp. 9.47-9.55 and the Pre-Gel Hybridization Technique taught in Vol. 4, No. 3 of PerSeptive Biosystems Magazine.
It is preferred that hybridization complexes be formed at a temperature of about 4°C to about 75°C for about 2 minutes to about 24 hours. It is particularly preferred to conduct denaturing for no more than 60 minutes in the presence of PNA probe, after which the temperature is passively cooled to room temperature without quenching.
It is possible to facilitate hybridization in solution by using certain reagents. Preferred examples of these reagents include single stranded binding proteins such as Rec A protein, T4 gene 32 protein, E. coli single stranded binding protein, major or minor nucleic acid groove binding proteins, divalent ions, polyvalent ions, and intercalating substances such as ethidium bromide, actinomycin D, psoralen, and angelicin.
The preferred markers for use in the invention are fluorophores. As will be appreciated by the skilled artisan, the wavelength preferably selected to induce fluorescence of the fluorescent marker is known in the art as the "excitation maximum," i.e., that wavelength which is absorbed by a molecule and excites that molecule to a higher electronic state. When the marker molecule passes from the higher to a lower electronic state, the molecule emits a type of visible radiation, i.e., fluorescence, at a wavelength referred to as the "emission maximum." It is at least this fluorescence that is detected in the present invention. The detectable signal emitted by the compound can be detected using techniques known in the art, for example by observation with the human eye, using electronic means for detecting a generated wavelength (e. g., cameras and CCDs), and the like. Advanta-geously, the wavelength of fluorescence is sufficiently removed from that of the exciting light to allow good separation of the two wavelengths by optical filters.
The excitation wavelength is selected (by routine 5 experimentation and/or conventional knowledge) to correspond to this excitation maximum for the marker being used, and is preferably 200 to 1000 nm. For example, when the marker is fluoroscein, the preferred wavelength of excitation is about 488 nm. Fluorescent dyes are preferably selected to 10 have an emission wavelength of 200 to 1000 nm.
In preferred embodiments, an argon ion laser is used to irradiate the marker with light having a wavelength in a range of 400 to 520 nm, and fluorescent emission is detected in a range of 500 to 750 nm.
An apparatus for performing the inventive method can comprise a liquid medium container for containing the liquid medium; a laser for irradiating the nucleotide; a CCD
fluorescence detector and/or photomultiplier for detecting fluorescence induced by the laser; a data analysis device for analyzing data generated by the fluorescence detector;
and an output device which reports the data analysis generated by the data analysis device. See, e.g., Fig. lA, which shows a schematic diagram of a f luorescence detection system suitable for use with the method of the invention.
Unlike certain prior art methods, no separation of the hybridization complexes from the uncomplexed probes is necessary in the present method. In certain prior art methods, unhybridized probes and hybridized probes must be separated to enhance the signal to noise ratio ( i. e. , the ratio of the hybridization complex signal to the unhybridized probe signal or noise) , enabling detection of hybridization. In the present method, the change in the overall signal is monitored without performing the additional burdensome step of separating the hybridized and unhybridized probes. The inventors have discovered that nucleotide sequence information can be determined by monitoring a change in the overall signal intensity, which is a function of hybridization and hybridiza-tion efficiency.
In particular, the inventors have discovered a signal quenching effect related to probe-nucleotide hybridization, wherein the intensity of laser induced fluorescence of an unbound probe exceeds that of the same probe bound to a nucleotide sequence. Therefore, a solution lacking any target sequences for probes therein will fluoresce more intensely than an otherwise identical solution containing target sequences and thus probe-nucleotide hybridization complexes.
Moreover, the intensity of laser induced fluorescence of hybridized probes is inversely proportional to the hybridization efficiency of the probes for their target sequences. Therefore, a solution containing imperfectly complementary target sequences for probes therein will fluoresce more intensely than an otherwise identical solution containing perfectly complementary target sequences. A
solution containing target sequences mismatching n bases of the probes therein will fluoresce more intensely than an otherwise identical solution containing target sequences mismatching less than n bases of the probes therein. Thus, a three mismatch solution fluoresces more intensely than a two mismatch solution, which fluoresces more intensely than a one mismatch solution, which fluoresces more intensely than a zero mismatch (completely complementary) solution.
The quenching effect can be used to obtain nucleotide sequence information in a variety of ways.
In embodiments, a predetermined amount of at least one PNA probe can be added to a predetermined volume of solution containing a predetermined amount of at least one nucleotide sequence to be detected. After subjecting the sample to hybridizing conditions, the sample's laser induced fluorescent intensity can be measured. Sequence information regarding the at least one nucleotide sequence in the sample can be determined by comparing the intensity with the intensity of at least one known sample against which the apparatus and the method are calibrated. Thus, a mutant form of the target sequence has been detected if, for example, (a) a sample containing DNA and PNA probes hybridizable to a sequence of wild type DNA, fluoresces significantly more intensely than (b) a standard sample containing a probe perfectly complemen-tary to the same sequence of the wild type DNA.
Similarly, a mutant form of the target sequence is detected if the sample does not fluoresce significantly less intensely than a standard sample containing a mutant form of the target sequence.
A significant difference in intensity is defined for present purposes as a difference not attributable to mere experimental variation. In the cases studied thus far, the intensity of a perfectly matched PNA probe and sequence has been at least about 40% lower than the intensity (at the same wavelength) of an imperfectly matched PNA probe and the same sequence. This value will vary along with the hybridization efficiency of a given case. However, those of ordinary skill in the art will readily appreciate that the actual value for any case being analyzed can be obtained empirically without undue experimentation.
The quenching effect appears to be most pronounced for DNA/RNA analog probes having uncharged backbones, such as PNA and methylene methyl amino oligonucleotides. The specification largely focuses on the most preferred embodiment of the invention -- assaying methods using PNA probes -- but the invention also encompasses the use of other antisense probes (DNA, DNA analog, RNA and/or RNA analog probes).
Another embodiment of the invention comprises dividing a sample into equal portions and treating each portion as a separate sample as discussed above, except that a different probe is added to each portion. The intensities of the portions are compared to determine, inter a3ia, which probe is most complementary, and thus which target sequence is in the original sample. This embodiment of the method is advantageous in that the system does not need to be calibrated against a known sample.
Although solid supports and gels are not required to practice this invention, such supports can be used in embodiments of the invention for purposes other than separating hybridization complexes from unhybridized probes. For example, two similar types of probes differing by one base can be fixed to opposing internal surfaces of a container in which a sample is added. After subjecting the sample to hybridization conditions and fluorescence inducing radiation, the fluorescent intensity emanating from the opposing surfaces of the container can be compared to determine whether the sample contains a nucleotide sequence perfectly complementary to either or both types of probes fixed to the surface.
A plurality of probes can be employed simultaneously to achieve a variety of effects. Several probes targeted for different segments of a single nucleotide sequence can be employed to enhance the reliability of the detection method.
Similarly, one probe can target one strand of dsDNA, while another probe can target the complementary strand of dsDNA.
A preferred method of detecting whether DNA is mutant type or the corresponding wild type comprises the simultaneous use of ( a ) a f first type of probe targeted to a sequence that occurs in both the wild type and mutant type DNA but is otherwise unique, and (b) a second type of probe targeted to a sequence unique to the mutant type DNA, wherein the f first and second types of probe have different markers that produce distinguishable signals. Thus, detection of the first probe signal indicates that the test was run properly (i.e., the first probe functions as a positive control) and detection of the second probe signal indicates that the mutant type DNA is present. For example, one probe can have a fluoresce in marker exhibiting a fluorescent emission intensity peak at 525 nm while the other probe can have a rhodamine marker exhibiting a fluorescent emission intensity peak at 580 nm.
The speed, accuracy and efficiency with which the invention is able to yield sequence data, coupled with the ability of laser induced fluorescence of probes to localize sequence portions within a longer sequence, make the invention an alternative to the FISH method for mapping genomes (compare, e.g., Heppell-Parton, "Gene Mapping by Fluorescence in Situ Hybridization," p. 350-54, in Molecu3ar Biology and Biotechnol-ogy: A Comprehensive Desk Reference (Myers, ed. 1995)).
The invention provides an efficient method for analyzing nucleotide conformation, which is particularly useful for designing antisense drugs. Antisense drugs typically target mRNA for hybridization with an antisense sequence, so as to prevent translation of the mRNA into undesirable proteins.
Unfortunately, the in situ folding of mRNA prevents some sequence portions along its length from being accessible to antisense sequences. Until now, drug designers have located accessible sequences by a method in which a series of antisense nucleotides complementary to different portions of the target mRNA are combined with the mRNA, and the best binding antisense nucleotide, presumably corresponding to the most accessible portion of the mRNA, is identified through. slow and laborious tissue culture experiments, which can take at least about 45 days. See, e.g., Rawls, "Optimistic About Antisense,"
75(22) Chem. Eng. News 35, 39 (June 2, 1997) . The accessible portions of mRNA can be identified using the instant invention without tissue culture experiments, since laser induced fluorescent intensity is inversely proportional to hybridiza-tion efficiency. The sequence emitting the lowest intensity has the highest hybridization efficiency with the target mRNA, and is presumably complementary to a segment that is not obstructed by in situ folding of mRNA.
The probes can be marked antisense drugs and/or can be analogs thereof. For example, it might be advantageous to design phosphorothioate oligonucleotide antisense sequences by performing laser induced fluorescent studies with such sequences marked with a fluorophore or with PNA probes similarly marked.
Moreover, the invention enables the length and other features of the antisense drugs to be readily fine tuned to optimize hybridization efficiency.

In contrast to prior art nucleotide sequence detection methods, the present invention makes it possible to limit the total volume of the liquid medium (i.e., the sample to be analyzed) in certain embodiments to about 5 microliters.
5 Typically, the total volume is about 5 microliters to about 2000 microliters.
i when testing for dsDNA using PNA, if a result is obtained for which there remains doubt, a further test may be immediately performed on the sample by adding the complementary 10 PNA probe to test the complementary strand of DNA.
Alternatively, the PNA test can be done with both the PNA
and complementary PNA probes hybridized to each of the denatured DNA strands in the first instance and at the same t ime .
15 For forensic applications, samples can be tested, stored and then retested because PNA is expelled from hybridization over a couple of days, and DNA recombines over time and does not degrade by this procedure. Accordingly, a sample frozen after testing can be subsequently retested in the same tube a number of times.
Clinical samples can be tested using at least 2000 times less chemicals or genomic material (5 microliters vs. 10 milliliters) than is typical in conventional- methods.
Therefore, even using 10 or 20 times the concentration of probe conventionally used, the tests still only consume 1/5th to 1/lOth the amount of probe, while obtaining a very decisive result.
The invention will be illustrated in more detail with reference to the following Examples, but it should be understood that the present invention is not deemed to be limited thereto.
EXAMPLES
The DNA used in the Examples was amplified by PCR and purified using the QIAquick*PCR Purification Kit (QIAGEN Inc. , Chatsworth, CA, USA). PCR was conducted using a GeneAmp*PCR
system 2400 from Perkin Elmer, with a hot start at 95°C for Trademark*
minutes. After adding Taq enzyme, 35 cycles were carried out as follows:
Denaturing: 94°C for 30 sees.
Annealing: 45°C for 45 sees. (45°C is 5°C lower 5 than the primer Tm) Extension: 72°C for 30 sees. (1 kb/min.) x 35.
PCR was conducted using a mixture comprising in 100 ~,1 volume 10 x reaction buffer (10 ~l), 20 mM of MgCl2 (10 ~,1), mM of dNTP mixture (2), 25 pmol/~1 of primer 1 (1 ~,l), 10 25 pmol/~1 of primer 2 (1 ~C1) , 10 ng to 100 mg/~1 of DNA
template (1 ~1) , dd HZO (74 ~cl) and Taq polymerase (1 ~1) (5 unit/~l).
The concentration of DNA in each sample was measured with a W spectrometer at 260 nm. Three related fragments of DNA were used in the Examples.
SEQ ID N0:1 was a 150 base pair fragment of genomic DNA
amplified and purified from wild type p53 DNA.
SEQ ID N0:2 was a 150 base pair fragment identical to the wild type fragment (SEQ ID N0:1) except for one base mutation at amino acid position 344 (bases 100-102) at which the wild type sequence CTG was changed to CAG.
SEQ ID N0:3 was a 150 base pair fragment identical to the wild type fragment (SEQ~ ID N0:1) except for one base mutation at amino acid position 344 (bases 100-102) at which the wild type sequence CTG was changed to CGG.
The PNA probes used in the Examples were synthesized by PerSeptiwe Biosystems, Inc. (Framingham, MA, USA).
Probe No. 1 was a 12-mer PNA probe designed to be completely complementary to a 12 nucleotide segment of the 150 by p53 wild type fragment (SEQ ID N0:1). The probe had the following structure:
5' H-Flu-O-CAT TCA GCT CTC Lys-CONHZ(SEQ ID N0:7).
Probe No. 2 was a 12-mer PNA probe designed to be completely complementary to a 12 nucleotide segment of SEQ
ID N0:2 and one a one base mismatch with SEQ ID NO:1. The probe had the following structure:
5~ H-Flu-O-CAT TCT GCT CTC Lys-CONH2(SEQ ID N0:8).
Probe No. 3 was a 12-mer PNA probe designed to be complementary to a 12 nucleotide segment of the 150 by p53 wild type fragment (SEQ ID N0:3) . The probe had the following structure:
5' H-Rho-O-CAT TCA GCT CTC Lys-CONH2(SEQ ID N0:9).
Prior to testing, the apparatus was calibrated against a known sample.
PNA probes in the examples were obtained from master solutions. Each 0.04 pmol/~el master solution was prepared by mixing 50 ~cl of a 1 pmol/~1 solution of the PNA probe in TFA (trifluoroacetic acid) and 1200 ~,1 0.5xTBE buffer to provide a PNA concentration of 0.04 pmol/~1. Each 0.05 pmol/~tl master solution was prepared by mixing 40 ~1 of a 1 pmol/~,1 solution of the PNA probe.in TFA (trifluoroacetic acid) and 760 ul 0. 5xTBE buffer .to provide a PNA concentration of 0. 05 pmol/~1. Each 0.10 pmol/~1 master solution was prepared by mixing 80 ~,l of a 1 pmol/~1 solution of the PNA probe in TFA
(trifluoroacetic acid) and 720 ~,1 0.5xTBE buffer to provide a PNA concentration of 0.10 pmol/~1. Each mixture was heated to 50°C, maintained at that temperature for 10 minutes and then cooled to room temperature over 20 minutes.
Fluorescence was induced and detected using the system schematically depicted in Fig. lA. An argon ion laser (Ion Laser Technology, Salt Lake City, UT, USA) was used along with an 52000 spectrometer (Ocean Optics, Inc. , Dunedin, FL, USA) . The wavelength of the laser beam produced by the laser was 488 nm.
F~XAMPLE lA ' 5 pmol of DNA (SEQ ID NO:1) was added into 125 ~1 of Probe No. 1 master solution (0.04 pmol PNA/ul) to provide a test solution comprising 5 pmol of SEQ ID NO:1, 5 pmol of Probe No. 1 and 120 ~1 0.5xTBE buffer. The sample was heated at 95°C for l0 minutes and hybridized at 25°C for 30 minutes.
After hybridization, the solution was immediately placed into a cuvette, irradiated with a laser beam and monitored for fluorescent emission.

Example lA was repeated with SEQ ID N0:2 substituted for SEQ ID NO:1.
Fig. 1B shows the fluorescence spectra obtained in Examples lA and 1B . The maximum f luorescent intensity occurred at about 525 nm, since the marker was fluorescein. The maximum intensity of the Example lA solution, containing DNA hybridized to completely complementary PNA probes, was about 40% lower than the maximum intensity of the Example 1B solution, containing DNA hybridized to incompletely complementary PNA
probes.

After the samples in Examples lA and 1B had been subjected to laser induced fluorescence and detection, the hybridization complexes in each were separated from the unhybridized probes in each by spinning at 700 rpm for 2 minutes in G50 spin columns. Unhybridized probes were retained on the columns, while the hybridization complexes filtered through the columns and were collected in a cuvette for further laser induced fluorescence detection.
Fig. 1C shows the resulting fluorescence spectra. The trend is the opposite of that shown in Fig. 1B. That is, the perfectly matched PNA-DNA sample of Example lA fluoresces more intensely than the imperfectly matched PNA-DNA sample of Example 1B. This is evidence that the hybridization yield in Example lA is much higher than that in Example 1B.

4 pmal of SEQ ID NO:1 was added into 80 ~.1 of Probe No. 1 master solution (0.05 pmol PNA/~.1) to provide a test solution comprising 4 pmol of SEQ ID NO:1, 4 pmol of Probe No. 1 and 76 ~,1 0.5xTBE buffer. The sample was heated at 95°C for 10 minutes and hybridized at 25°C for 30 minutes. After hybridization, the solution was immediately placed into a cuvette, irradiated with a laser beam and monitored for fluorescent emission.
Example 2A was repeated with 3 pmol of SEQ ID NO:1 and 1 pmol of SEQ ID N0:2 substituted for 4 pmol of SEQ ID N0:1.

Example 2A was repeated with 2 pmol of SEQ ID NO:1 and 2 pmol of SEQ ID N0:2 substituted for 4 pmol of SEQ ID N0:1.

Example 2A was repeated with 1 pmol of SEQ ID N0:1 and 3 pmol of SEQ ID N0:2 substituted for 4 pmol of SEQ ID NO:1.

Example 2A was repeated with 4 pmol of SEQ ID N0:2 substituted for 4 pmol of SEQ ID NO:1.
Fig. 2 shows the fluorescence spectra of Examples 2A-2E.
Example 2A, having the highest percentage of perfect complements, yielded the lowest intensity, while Example 2E, having the lowest percentage of perfect complements, yielded the highest intensity. The fluorescent intensities decrease with increasing concentration of PNA-DNA hybridization complexes.

8 pmol of DNA (SEQ ID NO:1) was added into 80 ~C1 of Probe No. 2 master solution (0.10 pmol PNA/ul) to provide a test solution comprising 8 pmol of SEQ ID NO:1, 8 pmol of Probe No. 2 and 72 ~cl 0.5xTBE buffer. The sample was heated at 95°C for 10 minutes and hybridized at 25°C for 30 minutes.
After hybridization, the solution was immediately placed into a cuvette, irradiated with a laser beam and monitored for fluorescent emission.

Example 3A was repeated with SEQ ID N0:2 substituted for SEQ ID NO:1.

Example 3A was repeated with SEQ ID N0:3 substituted for SEQ ID NO:1.
Fig. 3 shows the fluorescence spectra of Examples 3A-3C.
The intensity of Example 3B, in which a completely complemen-tary PNA probe and target sequence were combined, was significantly lower than the intensities of Examples 3A and 3C, in which incompletely complementary probes and targets were combined. The difference in intensities between Examples 3A and 3C is attributable to the type of mismatch between 5 the respective probes and target sequences. Example 3C had an unfavorable G-T mismatch, while Example 3A had an even less favorable T-T mismatch, and thus Example 3A had the lowest hybridization efficiency and the highest intensity.

10 4 pmol of SEQ ID N0:1 was added into 80 ~C1 of Probe No. 3 master solution (0.10 pmol PNA/~,1) to provide a test solution comprising 4 pmol of SEQ ID N0:1, 8 pmol of Probe No. 3 and 72 ~,1 0.5xTBE buffer. The sample was heated at 95°C for 10 minutes and hybridized at 25°C for 30 minutes. After 15 hybridization, the solution was immediately placed into a cuvette, irradiated with a laser beam and monitored for fluorescent emission.

Example 4A was repeated with SEQ ID N0:2 substituted 20 for SEQ ID NO:1.
Fig. 4 shows the fluorescence spectra obtained in Examples 4A and 4B. The maximum fluorescent intensity occurred at about 580 nm, since the marker was rhodamine. The maximum intensity of the Example 4A solution, containing DNA hybridized to completely complementary PNA probes, was about 40% lower than the maximum intensity of the Example 4B solution, containing DNA hybridized to incompletel~~ complementary PNA
probes.

Fig. 5 is a schematic diagram of the fluorescent detection system used in this example, including an argon ion laser (Ion Laser Technology) as a light source and a CCD array (Princeton Instruments, Inc. , Trenton, NJ, USA) as a detector.
4 pmol of SEQ ID NO:1 was added into 80 ~1 of Probe No.
1 master solution (0.05 pmol PNA/ul) to provide a test solution comprising 4 pmol of SEQ ID N0:1, 4 pmol of Probe No. 1 and 76 ~Cl 0.5xTBE buffer. The sample was heated at 95°C for IO
minutes and hybridized at 25°C for 30 minutes. After hybridization, the solution was immediately placed into a cuvette, irradiated with a laser beam and monitored for fluorescent emission.

Example 5A was repeated with SEQ ID N0:3, which is a one base mismatch with Probe No. 1, substituted for SEQ ID
NO:1.

Example 5A was repeated with SEQ ID N0:4, which is a two base mismatch with Probe No. 1, substituted for SEQ ID
NO:1.

Example 5A was repeated with SEQ ID N0:5, which is a three base mismatch with Probe No. 1, substituted for SEQ
ID NO:1.

Example 5A was repeated with SEQ ID N0:6, which lacks a target sequence for Probe No. 1, substituted for SEQ ID
NO:1.
Fig. 6 shows the spectra obtained in Examples 5A-5E.
The non-target sequence solution (5E) fluoresced more intensely than the three mismatch solution (5D) , which fluoresced more intensely than the two mismatch solution ( 5C) , which f luoresced more intensely than the one mismatch solution (5B), which fluoresced more intensely than the zero mismatch (completely complementary) solution (SA).

4 pmol of SEQ ID NO:1 was added into 80 ~,1 of Probe No.
4 (5' Fluo-CAT TCA GCT CTC 3') master solution (0.10 pmol DNA/~,1) to provide a test solution comprising 4 pmol of SEQ
ID NO:1, 8 pmol of Probe No. 4 and 72 ~,1 0.5xTBE buffer.
Probe No. 4 was a DNA probe designed to be completely complementary to a 12 nucleotide segment of SEQ ID NO:1.
The sample was heated at 95°C for 10 minutes and hybridized at 25°C for 30 minutes. After hybridization, the solution was immediately placed into a cuvette, irradiated with a laser beam and monitored for fluorescent emission using the system schematically depicted in Fig. 5 and described in Example 5A.

Example 6A was repeated with SEQ ID N0:2 substituted for SEQ ID N0:1.
Figs. 7A and 7B show the spectra obtained from Examples 6A and 6B, respectively. The fluorescent intensity of Example 6A (completely complementary) was slightly lower than that of Example 6B (one base pair mismatch).
1~ EXAMPLE 6C
After the samples in Examples 6A and 6B had been subjected to laser induced fluorescence and detection, any hybridization complexes in each were separated from the unhybridized probes in each by spinning at 700 rpm for 2 minutes in G50 spin columns. Unhybridized probes were retained on the columns, while any hybridization complexes would have filtered through the columns and been collected in a cuvette for further laser induced fluorescence detection.
Figs. 8A and 8B show the resulting fluorescence spectra.
There was no evidence of hybridization complexes.
A~tiVDED SHEET
IPEA/EP ~
(1) GENERAL INFORMATION
SEQUENCE LISTING
(i) APPLICANT:
(A) NAME: Lorne Park Research, Inc.
(B) STREET: 20 Queen Street West, Suite 2018, Box 101 (C) CITY: Toronto (D) STATE: Ontario (E) COUNTRY: Canada (F) POSTAL CODE (ZIP): M5H 3R3 (ii) TITLE OF INVENTION: Assaying Nucleotides in Solution using a Fluorescent Intensity Quenching Effect (iii) NUMBER OF SEQUENCES: 9 (iv) CORRESPONDENCE ADDRESS
(A) NAME: COWLING LAFLEUR HENDERSON LLP
(B) STREET: 160 ELGIN STREET, SUITE 2600 (C) CITY: OTTAWA
(D) PROVINCE: ONTARIO
(E) COUNTRY: CANADA
(F) POSTAL CODE: K1P 1C3 (v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: PatentIn Release #1.0, Version #1.30 (EPO) (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER: 2,295,333 (B) FILING DATE: 30-June-1998 (vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER: US 08/886,280 (B) FILING DATE: 1-July-1997 (viii) ATTORNEY/AGENT INFORMATION
(A) NAME: COWLING LAFLEUR HENDERSON LLP
(B) REFERENCE NUMBER: 08-885767CA
(ix) TELECOMMUNICATION INFORMATION
(A) TELEPHONE: 613-233-1781 (B) TELEFAX: 613-563-9869 (2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 150 bases (B) TYPE: nucleotide (C) STRANDEDNESS: double-stranded (D) TOPOLOGY: linear (ii) MOLECULE TYPE: genomic DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:

(2) INFORMATION FOR SEQ ID N0:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 150 bases (B) TYPE: nucleotide (C) STRANDEDNESS: double-stranded (D) TOPOLOGY: linear (ii) MOLECULE TYPE: genomic DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:2:

(2) INFORMATION FOR SEQ ID N0:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 150 bases (B) TYPE: nucleotide (C) STRANDEDNESS: double-stranded (D) TOPOLOGY: linear (ii) MOLECULE TYPE: genomic DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:3:

(2) INFORMATION FOR SEQ ID N0:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 150 bases (B) TYPE: nucleotide (C) STRANDEDNESS: double-stranded (D) TOPOLOGY: linear (ii) MOLECULE TYPE: genomic DNA

(xi) SEQUENCE DESCRIPTION: SEQ ID N0:4:

(2) INFORMATION FOR SEQ ID N0:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 150 bases (B) TYPE: nucleotide (C) STRANDEDNESS: double-stranded (D) TOPOLOGY: linear (ii) MOLECULE TYPE: genomic DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:5:

(2) INFORMATION FOR SEQ ID N0:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 633 bases (B) TYPE: nucleotide (C) STRANDEDNESS: double-stranded (D) TOPOLOGY: linear (ii) MOLECULE TYPE: genomic DNA
(xi) SEQUENCE DESCRIPTION: SEQ ID N0:6:

25a (2) INFORMATION FOR SEQ ID N0:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 12 bases (B) TYPE: nucleotide (C) STRANDEDNESS; single-stranded (D) TOPOLOGY: linear (ii) MOLECULE TYPE: synthetic DNA
(ix) FEATURE
(D) OTHER INFORMATION: nucleotide segment of Probe No.l (xi) SEQUENCE DESCRIPTION: SEQ ID N0:7:

(2) INFORMATION FOR SEQ ID N0:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 12 bases (B) TYPE: nucleotide (C) STRANDEDNESS: single-stranded (D) TOPOLOGY: linear (ii) MOLECULE TYPE: synthetic DNA
(ix) FEATURE
(D) OTHER INFORMATION: nucleotide segment of Probe No.2 (xi) SEQUENCE DESCRIPTION: SEQ ID N0:8:

(2) INFORMATION FOR SEQ ID N0:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 12 bases (B) TYPE: nucleotide (C) STRANDEDNESS: single-stranded (D) TOPOLOGY: linear (ii) MOLECULE TYPE: synthetic DNA
(ix) FEATURE
(D) OTHER INFORMATION: nucleotide segment of Probe No.3 (xi) SEQUENCE DESCRIPTION: SEQ ID N0:9:

Claims (28)

WHAT IS CLAIMED IS:
1. A method for detecting at least, one single stranded or double stranded nucleotide sequence in a liquid medium, said method comprising:
providing said liquid medium comprising said at least one nucleotide sequence adding to said liquid medium at least one antisense probe to form at least one hybridization complex with said at least one nucleotide sequence, wherein said at least one probe comprises at least one marker; and detecting said at least one nucleotide sequence by detecting at least one signal that is inversely proportional to an amount of said at least one hybridization complex in said liquid medium and proportional to an amount of said at least one probe unhybridized in said liquid medium, wherein said method is conducted without separating unhybridized probes from said at least one hybridization complex prior to said signal detecting, without detecting a change in anisotropy or polarity, and without providing a signal quenching agent on said at least one probe or on said at least one nucleotide sequence.
2. The method for detecting at least one nucleotide sequence according to claim 1, wherein said at least one probe has an uncharged backbone.
3. The method for detecting at least one nucleotide sequence according to claim 2, wherein said at least one probe comprises a PNA sequence.
4. The method for detecting at least one nucleotide sequence according to claim 1, wherein said at least one marker includes a fluorescent dye and said at least one signal is at least one fluorescent emission generated by irradiating said at least one marker with a light source.
5. The method for detecting at least one nucleotide sequence according to claim 4, wherein said light source is an argon ion laser which irradiates said at least one marker with light having a wavelength of about 200 to about 1000 nm.
6. The method for detecting at least one nucleotide sequence according to claim 5, wherein said at least one fluorescent emission is detected in a range of 200 to 1000 nm.
7. The method for detecting at least one nucleotide sequence according to claim 6, wherein said at least one signal is at least one fluorescent emission intensity generated by irradiating said at least one hybridization complex with a laser.
8. The method for detecting at least one nucleotide sequence according to claim 7, wherein a laser induced fluorescent intensity maximum of said liquid medium is compared with a laser induced fluorescent intensity maximum of a standard sample to detect whether said at least one nucleotide sequence is present in said liquid medium.
9. The method for detecting at least one nucleotide sequence according to claim 8, wherein a fluorescent intensity quenching effect correlated with probe to nucleotide sequence hybridization efficiency is analyzed to detect said at least one nucleotide sequence.
10. The method for detecting at least one nucleotide sequence according to claim 9 , wherein a f first said at least one probe is complementary to a first segment of a first nucleotide sequence, a second said at least one probe is complementary to a second segment of a second nucleotide sequence, and said first and second segments differ from each other.
11. The method for detecting at least one nucleotide sequence according to claim 10, wherein said first and second probes are completely complementary to said first and second segments, respectively.
12. The method for detecting at least one nucleotide sequence according to claim 11, wherein said first nucleotide sequence is complementary to said second nucleotide sequence.
13. The method for detecting at least one nucleotide sequence according to claim 12, wherein said first nucleotide sequence and said second nucleotide sequence are within opposing strands of double stranded DNA.
14. The method for detecting at least one nucleotide sequence according to claim 11, wherein said first nucleotide sequence and said second nucleotide sequence are in different genes.
15. The method for detecting at least one nucleotide sequence according to claim 14, wherein said first probe has a first marker which has a first fluorescent emission intensity at a first wavelength, said second probe has a second marker which has a second fluorescent emission intensity at a second wavelength, said first and second wavelengths are different, said first nucleotide sequence is detected by monitoring fluorescent emission intensity at said first wavelength and said second nucleotide sequence is detected by monitoring fluorescent emission intensity at said second wavelength.
16. The method for detecting at least one nucleotide sequence according to claim 11, wherein said first probe has a first marker which has a first fluorescent emission intensity at a first wavelength, said second probe has a second marker which has a second fluorescent emission intensity at a second wavelength, said first and second wavelengths are different, said first nucleotide sequence is detected by monitoring fluorescent emission intensity at said first wavelength and said second nucleotide sequence is detected by monitoring fluorescent emission intensity at said second wavelength.
17. The method for detecting at least one nucleotide sequence according to claim 16, wherein said first nucleotide sequence is a positive control expected to be present in said liquid medium being analyzed, and said second nucleotide sequence is detected by comparing said first intensity and said second intensity.
18. The method for detecting at least one nucleotide sequence according to claim 17, wherein the second nucleotide sequence is found only in a mutant genomic DNA and the first nucleotide sequence is found in said mutant genomic DNA and in a corresponding wild type genomic DNA.
19. The method for detecting at least one nucleotide sequence according to claim 18, wherein the first and second markers are selected from the group consisting of fluoresce in and rhodamine.
20. The method for detecting at least one nucleotide sequence according to claim 17, wherein a third probe having a third marker which has a third fluorescent emission intensity at a third wavelength differing from the first and second wavelengths is added to said liquid medium as a negative control which is not expected to hybridize with any nucleotide sequence, and said second nucleotide sequence is detected by comparing the first, second and third intensities.
21. The method for detecting at least one nucleotide sequence according to claim 9, wherein prior to subjecting said liquid medium to hybridizing conditions, said liquid medium is divided into a plurality of equal portions with a different probe in each portion, and the laser induced fluorescent intensities of said portions are compared to determine which probe is most complementary to said at least one nucleotide sequence.
22. The method for detecting at least one nucleotide sequence according to claim. 9, wherein at least two different types of probes differing by at least one base are fixed to different internal surfaces of a container in which said liquid medium is added, and after subjecting the liquid medium to hybridization conditions and fluorescence inducing radiation, fluorescent intensities emanating from the different surfaces of the container are compared to determine whether the sample contains a nucleotide sequence perfectly complementary to either or both types of said probes fixed to the surface.
23. The method for detecting at least one nucleotide sequence according to claim 1, further comprising determining a hybridization efficiency of said at least one probe with respect to said at least one nucleotide sequence.
24. The method for detecting at least one nucleotide sequence according to claim 1, wherein said at least one nucleotide sequence is mRNA, and antisense drug accessible segments of said mRNA are identified by comparing the fluorescent intensities of a plurality of different probes for a plurality of different segments of said mRNA.
25. The method for detecting at least one nucleotide sequence according to claim 1, further comprising comparing the fluorescent intensities of a plurality of different probes for a single segment of said at least one nucleotide sequence to determine a number of base mismatches between each of said probes and said segment.
26. The method for detecting at least one nucleotide sequence according to claim 1, further comprising detecting locations of a plurality of nucleotide sequences within a genome to map said genome.
27. The method for detecting at least one nucleotide sequence according to claim 1, wherein said at least one marker is one fluorophore, and said one fluorophore is the only fluorophore on said at least one probe.
28. The method for detecting at least one nucleotide sequence according to claim 1, wherein said method is conducted without using enzymes to digest said probe.
CA002295333A 1997-07-01 1998-06-30 Assaying nucleotides in solution using a fluorescent intensity quenching effect Expired - Fee Related CA2295333C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US08/886,280 1997-07-01
US08/886,280 US5846729A (en) 1997-02-27 1997-07-01 Assaying nucleotides in solution using a fluorescent intensity quenching effect
PCT/IB1998/001016 WO1999001578A1 (en) 1997-07-01 1998-06-30 Assaying nucleotides in solution using a fluorescent intensity quenching effect

Publications (2)

Publication Number Publication Date
CA2295333A1 CA2295333A1 (en) 1999-01-14
CA2295333C true CA2295333C (en) 2005-03-01

Family

ID=25388774

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002295333A Expired - Fee Related CA2295333C (en) 1997-07-01 1998-06-30 Assaying nucleotides in solution using a fluorescent intensity quenching effect

Country Status (10)

Country Link
US (1) US5846729A (en)
EP (1) EP1002125A1 (en)
JP (1) JP2001510681A (en)
AR (1) AR016125A1 (en)
AU (1) AU730865B2 (en)
CA (1) CA2295333C (en)
IN (1) IN189178B (en)
TW (1) TWI235765B (en)
WO (1) WO1999001578A1 (en)
ZA (1) ZA985758B (en)

Families Citing this family (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6555692B1 (en) * 1996-02-26 2003-04-29 California Institute Of Technology Preparation and use of bifunctional molecules having DNA sequence binding specificity
SE9601318D0 (en) * 1996-04-04 1996-04-04 Pharmacia Biosensor Ab Method for nucleic acid analysis
US6046004A (en) * 1997-02-27 2000-04-04 Lorne Park Research, Inc. Solution hybridization of nucleic acids with antisense probes having modified backbones
US6251591B1 (en) * 1997-02-27 2001-06-26 Lorne Park Research, Inc. Quantitative method for detecting nucleotide concentration
US6485901B1 (en) 1997-10-27 2002-11-26 Boston Probes, Inc. Methods, kits and compositions pertaining to linear beacons
AU1366299A (en) 1997-10-27 1999-05-17 Boston Probes, Inc. Methods, kits and compositions pertaining to pna molecular beacons
US6361942B1 (en) * 1998-03-24 2002-03-26 Boston Probes, Inc. Method, kits and compositions pertaining to detection complexes
US6858390B2 (en) 1998-12-31 2005-02-22 Ingeneus Corporation Aptamers containing sequences of nucleic acid or nucleic acid analogues bound homologously, or in novel complexes
US6294333B1 (en) * 1998-12-31 2001-09-25 Ingeneus Corp. Fluorescent intensity assay for protein or peptide binding to nucleic acids
US6420115B1 (en) 1999-12-21 2002-07-16 Ingeneus Corporation Cation mediated triplex hybridization assay
US6656692B2 (en) * 1999-12-21 2003-12-02 Ingeneus Corporation Parallel or antiparallel, homologous or complementary binding of nucleic acids or analogues thereof to form duplex, triplex or quadruplex complexes
US6403313B1 (en) * 1999-12-21 2002-06-11 Ingeneus Corporation Fluorescent intensity assay for duplex and triplex nucleic acid hybridization solution utilizing fluorescent intercalators
DK1471926T3 (en) 1999-06-01 2011-02-28 Baylor College Medicine Compositions and Methods for Therapeutic Use of an Atonal-Associated Sequence
WO2000079009A2 (en) 1999-06-22 2000-12-28 Invitrogen Corporation Improved primers and methods for the detection and discrimination of nucleic acids
US6645733B1 (en) 1999-06-25 2003-11-11 Ingeneus Corporation Fluorescent intensity method for assaying binding between proteins or peptides
US6830902B1 (en) 1999-07-02 2004-12-14 Invitrogen Corporation Compositions and methods for enhanced sensitivity and specificity of nucleic acid synthesis
WO2001038585A2 (en) * 1999-11-24 2001-05-31 The Regents Of The University Of California Polymer arrays and methods of using labeled probe molecules to identify and quantify target molecule expression
US20030181412A1 (en) * 1999-12-21 2003-09-25 Ingeneus Corporation Method for modifying transcription and/or translation in an organism for therapeutic, prophylactic and/or analytic uses
US6924108B2 (en) * 1999-12-21 2005-08-02 Ingeneus Corporation Nucleic acid binding enhancement by conjugation with nucleotides, nucleosides, bases and/or their analogues
US6911536B1 (en) 1999-12-21 2005-06-28 Ingeneus Corporation Triplex and quadruplex catalytic hybridization
US7052844B2 (en) * 1999-12-21 2006-05-30 Ingeneus, Inc. Purification of DS-DNA using heteropolymeric capture probes and a triplex, quadruplex or homologous duplex binding mechanism
US7309569B2 (en) 1999-12-21 2007-12-18 Ingeneus, Inc. Parallel or antiparallel, homologous or complementary binding of nucleic acids or analogues thereof to form duplex, triplex or quadruplex complexes
US6927027B2 (en) 1999-12-21 2005-08-09 Ingeneus Corporation Nucleic acid multiplex formation
US6265170B1 (en) 2000-01-24 2001-07-24 Ingeneus Corporation Homogenous assay of duplex of triplex hybridization by means of multiple measurements under varied conditions
US20030170659A1 (en) * 2000-01-24 2003-09-11 Ingeneus Corporation Electrical treatment of binding media to encourage, discourage and/or study biopolymer binding
US7220541B2 (en) 2000-01-24 2007-05-22 Ingeneus, Inc. Homogeneous assay of biopolymer binding by means of multiple measurements under varied conditions
US6982147B2 (en) * 2000-01-24 2006-01-03 Ingeneus Corporation Apparatus for assaying biopolymer binding by means of multiple measurements under varied conditions
US6613524B1 (en) 2000-01-24 2003-09-02 Ingeneus Corporation Amperometric affinity assay and electrically stimulated complexes of nucleic acids
JP2003529376A (en) * 2000-04-03 2003-10-07 サイティック コーポレイション Detection and typing of human papillomavirus using PNA probes
US6936443B2 (en) * 2000-04-03 2005-08-30 Cytyc Corporation Detection and typing of human papillomavirus using PNA probes
US20060073530A1 (en) * 2001-08-15 2006-04-06 Olaf Schneewind Methods and compositions involving sortase B
US20030165859A1 (en) * 2001-10-23 2003-09-04 Invitrogen Corporation Primers and methods for the detection and discrimination of nucleic acids
DE10155031A1 (en) * 2001-11-09 2003-05-28 Friz Biochem Gmbh Fluorescence quenching for the detection of ligate-ligand association events in electric fields
US20030166282A1 (en) 2002-02-01 2003-09-04 David Brown High potency siRNAS for reducing the expression of target genes
EP1470148B1 (en) 2002-02-01 2012-07-18 Life Technologies Corporation Double-stranded oligonucleotides
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
US6818420B2 (en) 2002-02-27 2004-11-16 Biosource International, Inc. Methods of using FET labeled oligonucleotides that include a 3′-5′ exonuclease resistant quencher domain and compositions for practicing the same
US20060099224A1 (en) 2002-08-12 2006-05-11 David Kirn Methods and compositions concerning poxviruses and cancer
US20040180345A1 (en) * 2003-03-14 2004-09-16 Ingeneus Corporation Pre-incubation method to improve signal/noise ratio of nucleic acid assays
CA2581086C (en) 2004-09-14 2023-11-07 The Regents Of The University Of Colorado, A Body Corporate Method for treatment with bucindolol based on genetic targeting
BRPI0516955A (en) * 2004-09-24 2008-09-30 Ingeneus Inc genetic assay
JP2006254784A (en) * 2005-03-17 2006-09-28 Institute Of Physical & Chemical Research Method for decreasing nonspecific amplification from primer dimer
US8980246B2 (en) 2005-09-07 2015-03-17 Sillajen Biotherapeutics, Inc. Oncolytic vaccinia virus cancer therapy
JP2009507853A (en) 2005-09-07 2009-02-26 ジェンネレックス インコーポレイティッド Systemic treatment of metastatic and / or systemic disseminated cancer using GM-CSF expressing poxvirus
ATE378427T1 (en) * 2005-10-17 2007-11-15 Rina Netzwerk Rna Technologien METHOD FOR DETERMINING AN UNKNOWN PNS SEQENCE AND ITS USES
ES2551892T3 (en) 2006-09-15 2015-11-24 Ottawa Health Research Institute Oncolytic rhabdovirus
US8929630B2 (en) * 2009-03-27 2015-01-06 Life Technologies Corporation Systems and methods for assessing images
CN102686728B (en) 2009-06-29 2018-09-18 卢米耐克斯公司 Chimeric primers with hairpin conformation and its application method
US9409983B2 (en) 2009-07-23 2016-08-09 The Board Of Trustess Of The University Of Illinois Methods and compositions involving PBEF inhibitors for lung inflammation conditions and diseases
WO2011032088A1 (en) 2009-09-11 2011-03-17 Arca Biopharma, Inc. Polymorphisms in the pde3a gene
WO2011032180A1 (en) 2009-09-14 2011-03-17 Jennerex, Inc. Oncolytic vaccinia virus combination cancer therapy
CN102858959B (en) 2009-12-10 2016-02-24 渥太华医院研究院 Oncolytic rhabdovirus
ES2587191T3 (en) 2009-12-23 2016-10-21 Arca Biopharma, Inc. Methods and compositions for cardiovascular diseases and conditions
AU2012204467B2 (en) 2011-01-04 2016-08-18 Sillajen, Inc. Generation of antibodies to tumor antigens and generation of tumor specific complement dependent cytotoxicity by administration of oncolytic vaccinia virus
WO2012158238A2 (en) 2011-02-28 2012-11-22 University Of Iowa Research Foundation Anti-müllerian hormone changes in pregnancy and prediction ofadverse pregnancy outcomes and gender
CA2829300A1 (en) 2011-03-08 2012-09-13 King Abdullah University Of Science And Technology Molecular biomarker set for early detection of ovarian cancer
ES2627529T3 (en) 2011-06-08 2017-07-28 Children's Hospital Of Eastern Ontario Research Institute Inc. Compositions for glioblastoma treatment
RU2483049C2 (en) * 2011-07-21 2013-05-27 Открытое акционерное общество "Научно-исследовательский институт полимерных материалов" Method of producing lot of rocket solid-propellant multi-grain charges
WO2013036799A2 (en) 2011-09-09 2013-03-14 Fred Hutchinson Cancer Research Center Methods and compositions involving nkg2d inhibitors and cancer
WO2014116721A1 (en) 2013-01-22 2014-07-31 The Arizona Board Of Regents For And On Behalf Of Arizona State University Geminiviral vector for expression of rituximab
MX2015010783A (en) 2013-02-21 2016-06-21 Children S Hospital Of Eastern Ontario Res Inst Inc Vaccine composition.
US20150098940A1 (en) 2013-10-03 2015-04-09 Oklahoma Medical Research Foundation Biomarkers for Systemic Lupus Erythematosus Disease Activity, and Intensity and Flare
EP3224362A4 (en) 2014-11-26 2018-06-06 The Regents of The University of California Therapeutic compositions comprising transcription factors and methods of making and using the same
JP6937294B2 (en) 2015-09-16 2021-09-22 ぺタオミクス, インコーポレイテッド Methods and Compositions for Genome Target Enrichment and Selective DNA Sequencing
WO2018140606A1 (en) 2017-01-26 2018-08-02 Oklahoma Medical Research Foundation Biomarkers for systemic lupus erythematosus disease activity, and intensity and flare
WO2020081204A1 (en) 2018-10-18 2020-04-23 Oklahoma Medical Research Foundation Biomarkers for a systemic lupus erythematosus (sle) disease activity immune index that characterizes disease activity
EP3874273B1 (en) 2018-10-31 2024-03-27 Arizona Board of Regents on behalf of the University of Arizona Biomarkers and methods of use for radiation-induced lung injury

Family Cites Families (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4220450A (en) * 1978-04-05 1980-09-02 Syva Company Chemically induced fluorescence immunoassay
FR2558172B1 (en) * 1984-01-16 1986-06-13 Inst Nat Sante Rech Med PROBE CONTAINING MODIFIED NUCLEIC ACID AND RECOGNITION BY SPECIFIC ANTIBODIES AND USE OF SUCH PROBE AND THESE SPECIFIC ANTIBODIES TO DETECT AND CHARACTERIZE A HOMOLOGATED DNA SEQUENCE
US5217866A (en) * 1985-03-15 1993-06-08 Anti-Gene Development Group Polynucleotide assay reagent and method
EP0232967B1 (en) * 1986-01-10 1993-04-28 Amoco Corporation Competitive homogeneous assay
JPH05508323A (en) * 1991-04-11 1993-11-25 デイド、ベーリング、インコーポレイテッド Detection of DNA/RNA by fluorescence polarization
EP0539585A4 (en) * 1991-04-22 1993-09-01 Takata Corporation Surface-coated member
US5994056A (en) * 1991-05-02 1999-11-30 Roche Molecular Systems, Inc. Homogeneous methods for nucleic acid amplification and detection
US5539082A (en) * 1993-04-26 1996-07-23 Nielsen; Peter E. Peptide nucleic acids
US5332659A (en) * 1992-04-09 1994-07-26 The United States Of America As Represented By The Secretary Of The Navy Light emission-or absorbance-based binding assays for polynucleic acids
DE4216949C2 (en) * 1992-05-22 1997-07-24 Christoph Prof Dr Dr Cremer Non-enzymatic method for in situ hybridization on specific samples
US5674698A (en) * 1992-09-14 1997-10-07 Sri International Up-converting reporters for biological and other assays using laser excitation techniques
US5503980A (en) * 1992-11-06 1996-04-02 Trustees Of Boston University Positional sequencing by hybridization
US5445935A (en) * 1992-11-23 1995-08-29 Royer; Catherine A. Quantitative detection of macromolecules with fluorescent oligonucleotides
DE69333991T2 (en) * 1992-11-27 2006-12-14 Canon K.K. Method and probe for the detection of nucleic acids
US5538848A (en) * 1994-11-16 1996-07-23 Applied Biosystems Division, Perkin-Elmer Corp. Method for detecting nucleic acid amplification using self-quenching fluorescence probe
US5723288A (en) * 1994-05-06 1998-03-03 The University Of North Carolina At Chapel Hill Method of fluorescent detection of nucleic acids and cytoskeleton elements using bis-dicationic aryl furans, and kits useful therefor
US5747247A (en) * 1994-07-25 1998-05-05 The Regents Of The University Of California Spectroscopic helicase assay
ATE196322T1 (en) * 1995-05-05 2000-09-15 Perkin Elmer Corp METHODS AND REAGENTS FOR COMBINING PCR AMPLIFICATION WITH A HYBRIDIZATION ASSAY
DE69634696T2 (en) * 1995-10-06 2006-01-19 PerSeptive Biosystems, Inc., Framingham METHOD AND TEST SYSTEM FOR HYBRIDIZATION ANALYSIS USING PEPTIDE NUCLEIC ACID PROBLEMS
CA2190430A1 (en) * 1995-12-12 1997-06-13 Margret Barbara Basinski Method for measuring genetic messages

Also Published As

Publication number Publication date
AU730865B2 (en) 2001-03-15
AR016125A1 (en) 2001-06-20
CA2295333A1 (en) 1999-01-14
TWI235765B (en) 2005-07-11
EP1002125A1 (en) 2000-05-24
IN189178B (en) 2003-01-04
WO1999001578A1 (en) 1999-01-14
US5846729A (en) 1998-12-08
JP2001510681A (en) 2001-08-07
AU8030298A (en) 1999-01-25
ZA985758B (en) 1999-01-27

Similar Documents

Publication Publication Date Title
CA2295333C (en) Assaying nucleotides in solution using a fluorescent intensity quenching effect
US6046004A (en) Solution hybridization of nucleic acids with antisense probes having modified backbones
US6060242A (en) PNA diagnostic methods
EP0975800B1 (en) Assaying nucleotides in solution using pna probes
EP1078100B1 (en) Quantitative method for detecting nucleotide concentration
US6403313B1 (en) Fluorescent intensity assay for duplex and triplex nucleic acid hybridization solution utilizing fluorescent intercalators
US6821724B1 (en) Methods of genetic analysis using nucleic acid arrays
Ferguson-Smith Genetic analysis by chromosome sorting and painting: phylogenetic and diagnostic applications
US20060281102A1 (en) Methods for detecting genetic haplotypes by interaction with probes
KR20070011354A (en) Detection of strp, such as fragile x syndrome
US6844154B2 (en) High throughput methods for haplotyping
KR20040024588A (en) Homogeneous Assay of Biopolymer Binding by Means of Multiple Measurements Under Varied Conditions
US20030198983A1 (en) Methods of genetic analysis of human genes
RU2378383C1 (en) Set of synthetic oligonucleotides to detect dna of agent of vegetable desease - gram-negative bacterium pseudomonas syringae pv lachrymans by polymerase chain reaction
JP2004033003A (en) Label-free method for detecting variation in nucleic acid comprising single nucleotide polymorphism by using pna and single-stranded nucleic acid-cleaving nuclease

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed