throbber
(19) United States
`(12) Patent Application Publication (10) Pub. No.: US 2005/0142559 A1
`Makrigiorgos
`(43) Pub. Date:
`Jun. 30, 2005
`
`US 2005O142559A1
`
`(54) AMPLIFICATION OF DNA IN A HAIRPIN
`STRUCTURE, AND APPLICATIONS
`(75) Inventor: G. Mike Makrigiorgos, Brookline, MA
`(US)
`Correspondence Address:
`Ronald I. Eisenstein
`NXON PEABODY LLP
`100 Summer Street
`Boston, MA 02110 (US)
`(73) Assignee: Dana-Farber Cancer Institute, Inc.,
`Boston, MA (US)
`
`(21) Appl. No.:
`(22) Filed:
`
`10/758,401
`Jan. 15, 2004
`
`Related U.S. Application Data
`(60) Provisional application No. 60/440,184, filed on Jan.
`15, 2003.
`Publication Classification
`
`(51) Int. Cl. .............................. C12Q 1/68; C12P 19/34
`(52) U.S. Cl. ............................................... 435/6; 435/91.2
`
`(57)
`
`ABSTRACT
`
`The present invention is directed to a hairpin nucleic acid
`Structure and its use. In a preferred embodiment, the hairpin
`nucleic acid Structure can be used in a method of amplifi
`cation of a template nucleic acid Sequence that Substantially
`reduces polymerase-induced errors.
`
`Oxford, Exh. 1006, p. 1
`
`

`

`Patent Application Publication Jun. 30, 2005 Sheet 1 of 6
`
`US 2005/0142559 A1
`
`9
`
`
`
`EdÄL CITIWA
`
`1)T
`
`
`
`
`
`sueyuII pue deo e?efilT
`
`
`
`
`
`Oxford, Exh. 1006, p. 2
`
`

`

`Patent Application Publication Jun. 30, 2005 Sheet 2 of 6
`
`US 2005/0142559 A1
`
`?z ,
`
`gc og gø. Oz G! 0) ? ß
`
`SETOWO?Od9 g º £ Z !
`
`(Au) ONWSOS9W
`
`
`
`
`
`Oxford, Exh. 1006, p. 3
`
`

`

`Patent Application Publication Jun. 30, 2005 Sheet 3 of 6
`
`US 2005/0142559 A1
`
`3A
`
`Taq Site
`
`Alu site
`
`W
`Ligate Cap1 and Cap2
`in a single reaction
`Cap2
`
`v
`Cap1
`Treat W. uracil glycosylase.
`Add primers, start hairpin PCR
`
`
`
`DURING PCR OPO w
`
`Heating generates
`a strand break at the abasic site
`
`HARPIN SAMPLIFED
`
`Oxford, Exh. 1006, p. 4
`
`

`

`Patent Application Publication Jun. 30, 2005 Sheet 4 of 6
`
`US 2005/0142559 A1
`
`Figure 4A
`primer-binding site
`
`TN
`
`primer-binding site T
`
`Figure 4B
`primer-binding site
`
`primer-binding site
`
`Oxford, Exh. 1006, p. 5
`
`

`

`Patent Application Publication Jun. 30, 2005 Sheet 5 of 6
`
`US 2005/0142559 A1
`
`
`
`
`
`Oxford, Exh. 1006, p. 6
`
`

`

`Patent Application Publication Jun. 30, 2005 Sheet 6 of 6
`
`US 2005/0142559 A1
`
`A)
`S/
`N
`N)
`1A
`
`
`
`Oxford, Exh. 1006, p. 7
`
`

`

`US 2005/0142559 A1
`
`Jun. 30, 2005
`
`AMPLIFICATION OF DNA IN A HARPIN
`STRUCTURE, AND APPLICATIONS
`
`CROSS-REFERENCE TO RELATED
`APPLICATIONS
`0001) This application claims benefit under 35 U.S.C. S
`119(e) of U.S. Provisional Application No. 60/440,184, filed
`Jan. 15, 2003, which is incorporated herein by reference.
`
`FIELD OF THE INVENTION
`0002 The present invention is directed to a hairpin
`nucleic acid structures and its use. In a preferred embodi
`ment, the hairpin nucleic acid structure can be used in a
`method of amplification of a template nucleic acid Sequence
`that Substantially reduces polymerase-induced errors.
`
`BACKGROUND OF THE INVENTION
`Substantial interest has been directed to the detec
`0.003
`tion of changes in nucleic acid Sequences, Such as caused by
`mutation and methylation. For example, mutation in certain
`genes have been associated with a variety of disorders
`ranging from blood disorders to cancers. Genetic testing is
`one way to find this information out. However, our ability to
`detect Such mutations is limited by certain problems with a
`key component in these tests, namely the polymerase chain
`reaction (PCR).
`0004. A major problem with PCR is that polymerases
`invariably generate errors during amplification. Such poly
`merase misincorporations can be indistinguishable from
`genuine mutations, and lower the quality of DNA cloning
`and protein functional analysis by in Vitro translation. Poly
`merase misincorporations Set a limit for molecular mutation
`detection methods: the most Selective technologies invari
`ably rely on PCR, but PCR also poses a final selectivity
`limit, typically 1 mutant in 10-10° alleles, since all DNA
`polymerases generate errors during DNA Synthesis which
`can be misinterpreted as mutations (false positives). Thus,
`high Selectivity mutation detection technologies often fall
`Short of the enormous Selectivity needed to address issues
`like the generation of Spontaneous mutations in Somatic
`tissues '', the early detection of genomic instability, the
`mutation screening of single cells" or the reliable detection
`of minimal residual disease .
`. Both unknown and known
`mutation detection methods are affected by PCR errors and
`the most Selective methods are affected most.
`0005 For example, the principal limitation for mutation
`Scanning via constant denaturant capillary electrophoresis
`(CDCE) is the fidelity of the polymerase used 7 . High
`selectivity mutation scanning via DGGE and dHPLC is
`ultimately hindered by polymerase error rate ' ' '. Some of
`the highest sensitivity assays for RFLP-based known muta
`tion detection, including PCR/RE/LCR '', MutEx-ACB
`PCR , Radioactivity-based PCR-RFLP, RSM ' ',
`APRIL-ATM , and others reviewed in Parsons et al. 7,
`utilize PCR in at least one stage prior to RFLP-selection, and
`are therefore also limited by PCR errors .
`0006 Accordingly, it would be desirable if one had a
`means of amplifying DNA free of polymerase-induced mis
`incorporations, to detect mutations without being limited by
`polymerase-induced errors. This could significantly impact
`mutation detection, disease diagnosis, and cancer diagnosis.
`
`SUMMARY OF THE INVENTION
`0007 We have now discovered compositions and meth
`ods to amplify a target nucleic acid Sequence, Sometimes
`referred to as the template, that Substantially reduces poly
`merase induced errors in a Sequence of interest, and which
`can Supply existing technologies with the necessary Selec
`tivity leap. The first step of this method involves converting
`the Sequence of interest into a hairpin, which contains a
`double Stranded region linked at one end through a single
`Stranded loop, and performing PCR on the hairpin-Structure.
`In the second step, the amplified PCR products are heat
`denatured and rapidly cooled, to convert each amplified
`PCR product into a hairpin: genuine polymorphisms or
`mutations will remain fully matched in the hairpin, whereas
`PCR products which contain a PCR induced error will form
`a hairpin that contains a mismatch in the double-Stranded
`region. Thereafter, one removes those amplified nucleic
`acids which contain a mismatch by Standard means. This
`method results in an amplified target nucleic acid which is
`Substantially free of polymerase induced errors.
`0008. In an alternative embodiment, amplification of the
`hairpin Structure is performed using isothermal rolling circle
`amplification (RCA).
`0009 True nucleic acid changes such as from a mutation
`can be separated from polymerase-generated Single nucle
`otide changes, insertions, deletions, or Slippage thereby
`providing practically error-leSS nucleic acid, preferably
`DNA. By using a hairpin sequence one can obtain a sample
`(template) from a range of Sources Such as from genomic
`DNA. Large fractions of the human genome can be ampli
`fied via hairpin PCR to provide faithfully-replicated
`genomic DNA for extensive, genome-wide Screening for
`differences from a standard. This is particularly desirable
`when Starting from limited amounts of biopsy material, i.e.
`from a few cells obtained via laser capture microdissection.
`0010 Additional technical factors limit the overall selec
`tivity of mutation detection (e.g. amount of DNA, mis
`priming; heteroduplex formation; incomplete enzymatic
`digestion'); however, with appropriate selection of condi
`tions these problems can often be overcome. In contrast,
`PCR errors have been regarded as a glass ceiling for
`mutation detection Selectivity. The present method of using
`hairpin PCR will allow a boost to almost every existing
`method for highly Selective mutation detection and lead to
`Studies and diagnostic tests that were impossible with pre
`vious technology by Substantially reducing the number of
`errors that are an artifact of PCR from the sample. This
`method will also improve microSatellite analysis by elimi
`nating polymerase slippage artifacts 19 and will also have
`application in other areas Such as molecular beacons ''f''
`and real time PCR, DNA cloning for protein functional
`analysis by in vitro translation ".
`0011. In one embodiment of the present invention, a
`hairpin with non-complementary ends can be efficiently
`PCR-amplified. In this embodiment, a target DNA sequence
`which needs to be PCR-amplified is first converted to a
`hairpin following ligation of an oligonucleotide cap on one
`end and a pair of non-complementary linkers on the other
`end (See FIG. 1A). Next, primers corresponding to the two
`non-complementary linkers are used in a PCR reaction that
`proceeds by displacing the opposite Strand and amplifying
`the entire complement of the hairpin.
`
`Oxford, Exh. 1006, p. 8
`
`

`

`US 2005/0142559 A1
`
`Jun. 30, 2005
`
`0012. In one preferred embodiment, these primers corre
`sponding to the non-complementary linkers can overlap the
`Sequence of interest, thus conferring Sequence Specificity. In
`this embodiment, exponential PCR amplification of the
`hairpin is enabled and Sequences can be amplified directly
`from human genomic DNA. Following hairpin amplifica
`tion, the PCR product is heat-denatured to allow the hairpins
`to Separate from their complementary Strand, and placed
`rapidly on ice. Because of the Sudden cooling, cross-hybrid
`ization of different hairpins is minimal, and thus the original
`hairpins are reformed, following their amplification.
`0013 By amplifying DNA in a hairpin-formation, poly
`merase-errors practically always end-up forming a mis
`match. Genuine mutations, however, remain fully-matched.
`For example, if the polymerase introduces an A>G mutation
`on the upper Strand of the original Sequence, it is very
`unlikely that, during Synthesis of the bottom Strand of a
`Single hairpin it will perform the exact opposite error (TYC
`mutation) at exactly the complementary-Strand position.
`This can be seen when one looks at the normal probability
`for Such a double-error. Even for a polymerase with a large
`error rate of 10"/base the odds for a double-error event are
`10'x10'x0.25=2.5x10, i.e. less than the expected spon
`taneous mutation rate in somatic tissues '''. On the other
`hand, practically all genuine mutations remain fully matched
`following hairpin-PCR, as these reside in both strands from
`the beginning (FIG. 1A).
`0014 Preferably, the amplified hairpins that contain mis
`matches are efficiently Separated from those that do not,
`using any procedure that recognizes mismatch. Preferred
`methods include dHPLC-mediated fraction collection and
`enzymatic based Separation. Preferably, the hairpin caps are
`removed Subsequent to the Separation of hairpins containing
`mismatches from mismatch-free hairpins, thus allowing the
`original DNA sequence to be recovered. While the amplified
`DNA will have PCR-induced errors Such errors can be
`removed from the amplified Sample, which can now be
`processed for mutation detection without Sensitivity being
`limited by polymerase errors.
`0015. In a further preferred embodiment, DGGE,
`dHPLC, as well as methods based on the mismatch-binding
`protein MutS or Ce1I or resolvases (endo V) or exomu
`cleases are used to separate the fraction of PCR-amplified
`sequences containing polymerase errors 7 '
`'. These
`methods utilize the conversion of homoduplexes to hetero
`duplexes via cross-hybridization of PCR amplified products.
`Previously, both mutations and PCR errors are simulta
`neously converted to mismatches. When mutations are at a
`low frequency, practically all of them are converted to
`mismatches. Thus, Such a means did not discriminate them
`from PCR errors. By the present method mutations and other
`preexisting changes do not appear as mismatches. The
`present method of using a hairpin Structure takes advantage
`of the fact that genuine mutations are witnessed in both
`upper and lower DNA strands while PCR errors occur on
`one Strand at a time. Forcing DNA polymerase to copy both
`Strands in one pass creates a double record of the Sequence.
`Thus, effectively the method boosts the replication fidelity
`and converts PCR errors, but not other changes to mis
`matches.
`0016. The method of the present invention has wide
`applicability. For example, polymerase Slippage errors pro
`
`duce Stutter banding that complicate microSatellite analysis
`of single ', or pooled samples . Scanning for very low
`frequency changes occurring naturally in Somatic tissues (<1
`mutant in 107 alleles, ) or at early stages of carcinogenesis
`will enable identification of tumor Signatures as markers for
`early tumor detection. Identification of low level mutations
`in Somatic tissues will also facilitate elucidation of carcino
`gen-Specific mutational fingerprints following environmen
`tal exposures '. Reliable Screening for traces of 'onco
`mutations' can enhance the clinical and diagnostic
`utility of minimal residual disease detection
`and the
`identification of mutations in bodily excretions
`. For
`investigating the mechanisms of carcinogenesis, determina
`tion of carcinogen-induced mutational spectra in disease
`related genes in non-tumorous tissues can provide evidence
`as to whether a specific mutagenic agent or pathway is
`involved in a particular disease or cancer. This high-Selec
`tivity mutational spectrometry will also help determine
`whether or not a mutator phenotype must be invoked to
`explain the acquisition of multiple mutations in tumor cells
`18.32
`
`0017 Most previous studies of mutational spectra were
`based on phenotypic selection methods (e.g. HPRT, lacz
`assays). These methods preclude analysis of genes and
`human tissues for which Selective conditions cannot be
`devised in in-vitro Single cell Systems. Molecular methods
`with Selectivity comparable to the Spontaneous mutation
`frequency (107-10) that can be applied to all tissues are
`highly desirable
`7. However, the onset of PCR errors
`limits several approaches, such as CDCE, which would
`otherwise have the Sensitivity needed to measure the Spon
`taneous mutation frequency .
`0018) Mutation scanning methods such as DGGE or
`dHPLC are particularly hampered by PCR errors since, by
`detecting all possible mutations, they are more likely than
`RFLP-based methods to encounter misincorporation
`hotspots which result in false positives. Particularly for
`mutation detection from limited Starting material, Such as
`micrometastatic cells or laser capture microdissected
`Samples, very large DNA amplification is required. The error
`rate of conventional PCR is then particularly problematic
`as error containing Sequences can comprise >30% of the
`overall population ', making it almost impossible to iden
`tify genuine mutations. The present method changes that and
`it allows, for example dHPLC to overcome PCR errors and
`to perform reliable mutation analysis when Starting from a
`few cells or from minute, laser capture microdissected
`specimens. RFLP-based methods can now be used to exam
`ine few sites for mutations relative to mutation Scanning
`methods.
`0019. When a sample is limited, such as in minute
`LCM-dissected Samples, it previously was often not possible
`to perform more than a single PCR amplification towards the
`detection of mutations in one gene. With the present method,
`one can now perform mutation Screening in Several genes
`Simultaneously from a single Sample, for disease gene
`discovery or diagnostic applications. This whole genome
`amplification method permits amplification of genomic
`DNA from small tissue samples in an error-free manner. This
`allows repeated multi-gene mutation Screening from large
`collections of minute fresh or paraffin-embedded Samples
`without being limited by available starting material or PCR
`COS.
`
`Oxford, Exh. 1006, p. 9
`
`

`

`US 2005/0142559 A1
`
`Jun. 30, 2005
`
`0020) By removing PCR errors from amplified
`sequences, the present hairpin-PCR permits the use of
`well-established techniques such as dHPLC, CDCE, RFLP
`and microsatellite analysis for detecting traces of mutations
`in minute biopsies and for investigating the origins of cancer
`in human tissues without the introduction of polymerase
`induced errors.
`BRIEF DESCRIPTION OF THE DRAWINGS
`0021 FIGS. 1A-1C outline the generation of error-free
`amplified DNA via hairpin PCR. In FIG. 1A, the scheme for
`removing PCR errors following amplification of DNA in a
`hairpin structure is shown. FIG. 1B shows the expected
`structure and sequence of hairpin A. FIG. 1C shows the
`expected structure and sequence of hairpin D, an oligonucle
`otide encompassing both top and bottom Strands of p53 exon
`9.
`0022 FIGS. 2A-2H show PCR amplification and
`dHPLC separation of hairpin-shaped oligonucleotides. FIG.
`2A, lanes 1-5 show the PCR product of hairpins A,B,C,E,
`and D, respectively. Lanes 6 and 7 of FIG. 2A show
`amplification of hairpin D with only forward or only reverse
`primer. FIG. 2B shows amplification of hairpin C using
`Advantage Titanium(8) (lane 1), Pfu Turbo6 (lane 3) or
`Advantage HF2(R) (lane 5) polymerases respectively; lanes
`2, 4 and 6 are water-controls (no template) in each case.
`FIG. 2C shows quantitative real time PCR of hairpin D:
`curves 1-4, starting material of 1 ng, 100 pg, 10 pg and 1 pg
`respectively. FIG. 2D shows hairpin PCR (lanes 1 and 2, in
`duplicate) followed by denaturation and rapid cooling of the
`product (lanes 3 and 4, in duplicate). FIG.2E shows hairpin
`D amplified with primers that bind the non-complementary
`ends, and either not extending (lane 1) or extending 9 bases
`into the hairpin sequence (lane 2). FIG.2F shows spiking of
`p53 exon 9-containing hairpin D into 100 ng p53-negative
`HL-60 genome, followed by hairpin PCR using Advantage
`Titanium(R) polymerase. Spiking of 0.01 pg hairpin D cor
`responds to adding a single p53 exon 9 allele in the genome.
`Lanes 1-6, hairpin D addition of 0, 0.1, 1, 10, 100, 1000 pg
`respectively. FIG. 2G is similar to FIG. 2F, but using
`Advantage HF2(R) polymerase. Lanes 1-5, hairpin Daddition
`of 0, 0.01, 0.1, 1, 10 pg. FIG. 2H shows dHPLC-based
`separation of 1:1 mixtures of homoduplex and heteroduplex
`hairpins. The threshold of the fraction collector is set on the
`trailing (slowest) portion of the homoduplex.
`0023 FIGS. 3A-3B show conversion of a DNA sequence
`to a hairpin and PCR amplification. FIG. 3A shows the
`procedure used to convert a native DNA sequence, flanked
`by two different restriction sites, into a hairpin with non
`complementary ends that can be amplified. The hairpin
`shaped oligonucleotides Cap1 and Cap2 are ligated to the 5'
`and 3' of both sequence ends. During hairpin PCR, primers
`extending into the sequence are used to confer sequence
`specificity. FIG. 3B shows conversion of a native p53
`sequence flanked by Taq I/Alu I Sites to a hairpin, followed
`by hairpin-PCR. Lane 1: Hairpin-PCR product obtained by
`applying the scheme in FIG. 1A for an isolated p53
`sequence. Lane 2: Hairpin-PCR product obtained by apply
`ing the scheme in FIG. 1A to human genomic DNA, in order
`to directly amplify the same Alu I/Taq I target Sequence
`depicted in Lane 1. Lane 3: AS in lane 2, but omitting the
`addition of ligase from scheme FIG. 1A. Lanes 4 and 5: AS
`in lane 2, but omitting the forward or reverse primer,
`respectively, from PCR.
`
`0024 FIGS. 4A and 4B depict two preferred DNA
`structures. FIG. 4A depicts a DNA structure with a hairpin
`at one and non-complementary ends at the other end. FIG.
`4B depicts a DNA structure with hairpins at both ends of the
`double-stranded DNA.
`0.025 FIG.5 depicts the use of hairpin-shaped DNA as a
`detector for radiation and/or chemical exposures. The DNA
`strand breaks off following a strand break anywhere in the
`shaded area (target), thereby allowing the primers to bind
`and to PCR amplify the DNA segment. The amount of PCR
`amplification is proportional to how many DNA molecules
`undergo strand breaks and therefore it can be used to
`quantify the amount of radiation or chemical agent interact
`ing with the DNA. Finally, the fraction of DNA molecules
`that remain intact can be re-amplified by using primers
`binding to the non-complementary linkers, thereby regen
`erating the original DNA detector molecule.
`0026 FIG. 6 shows amplification of hairpins using roll
`ing-circle amplification (RCA). The hairpin-shaped oligo
`nucleotide of FIG. 6A was self-ligated to form a closed
`“dumbbell-like structure resembling the structures used for
`RNA-interference. The dumbbell was then amplified in an
`isothermal rolling-circle amplification reaction using Phi29
`polymerase (from New England Biolabs) and random prim
`ers. Following digestion of the RCA product with Alu, the
`amplified hairpin-dimer DNA was recovered. FIG. 6B
`shows in lane 1, no Alu digestion; in lane 2, digestion with
`Alu. The amplification is about 1000-fold. In another
`example, the hairpin-shaped oligonucleotide of FIG. 6C
`was self-ligated to form a closed 'dumbbell-like structure,
`and then amplified in an isothermal rolling-circle amplifi
`cation reaction using Phi29 polymerase (from New England
`Biolabs) and random primers. Following digestion of the
`RCA product with Nla-III, the amplified hairpin-dimer DNA
`was recovered. FIG. 6D shows in lane 1, no digestion
`Nla-III; lane 1: with Nla-III digestion). The amplification is
`about 500-fold.
`
`DETAILED DESCRIPTION OF THE
`INVENTION
`0027) We have discovered compositions and a method to
`amplify a target nucleic acid sequence, Sometimes referred
`to as the template, that Substantially reduces polymerase
`induced errors in a sequence of interest, and which can
`supply existing technologies with the necessary selectivity
`leap'. The first step of the method involves converting the
`sequence of interest into a hairpin, which contains a double
`stranded region linked at one end through a single Stranded
`loop, and performing PCR on the hairpin-structure. In the
`second step, the amplified PCR products are heat denatured
`and rapidly cooled, to convert each amplified PCR product
`into a hairpin: genuine polymorphisms or mutations will
`remain fully matched in the hairpin, whereas PCR products
`which contain a PCR induced error will form a hairpin that
`contains a mismatch in the double-stranded region. There
`after, one removes those amplified nucleic acids which
`contain a mismatch by standard means. This method results
`in an amplified target nucleic acid which is Substantially free
`of polymerase induced errors.
`0028) Any method of converting the nucleic acid to a
`hairpin with non-complementary ends can be used. AS used
`herein, hairpin structures include hairpins and dumbbells.
`
`Oxford, Exh. 1006, p. 10
`
`

`

`US 2005/0142559 A1
`
`Jun. 30, 2005
`
`Preferably, one uses oligonucleotide caps which, in a
`Single ligation Step allow the conversion of a native DNA
`Sequence to a hairpin with non-complementary ends. For
`example, one transforms the template nucleic acid, prefer
`ably DNA, into a hairpin by capping it at one end, Cap1.
`Cap1 is Sometimes referred to as a joining Structure,
`because once it is ligated to the nucleic acid Sequence of
`interest it joins the upper Strand of the nucleic acid Sequence
`of interest to the lower Strand of the Same nucleic acid
`molecule. Preferably, one caps the template at the other end,
`Cap2. Cap 2 is Sometimes referred to as a priming structure,
`because it contains regions of Single-Stranded nucleic acid to
`which primers can bind to initiate the polymerization reac
`tion. CapS1 and 2 naturally form hairpins on their own, to
`allow their respective ligation to the double stranded DNA
`ends of the template DNA. In addition, Cap2 contains a
`region with two non-complementary Sequences to allow
`Subsequent primer binding. Finally, Cap2 contains a poly
`merase block approximately at the center. This block can
`be one or more Synthetic abasic Sites, or a deoxynucleotide
`analogue that does not allow polymerase Synthesis, or a
`uracil that, upon treatment with uracil glycosylase and
`heating is converted to a Strand break, thus effectively
`providing the polymerase block. Any of the above men
`tioned polymerase blocks will enable the formation of a
`hairpin with non-complementary ends during the Subse
`quent PCR amplification. See FIG. 4. Alternatively, Cap2 or
`the priming Structure can be a pair of oligonucleotides with
`are complementary to each other at the ends ligated to the
`nucleic acid of interest, and non-to each other at their other
`ends.
`0029. In one preferred embodiment of the present inven
`tion, unbalanced concentrations of primers are used during
`PCR (asymmetric PCR) such that the result of amplifica
`tion is a single Stranded nucleic acid product (i.e. monomer
`hairpins) instead of a double Stranded product (dimer hair
`pins). In this embodiment, denaturation-renaturation of the
`DNA is unnecessary.
`0.030. In contrast to the method developed by Jones et al.
`(Jones and Winistorfer, 1992) (“panhandle PCR) where the
`overall Structure is in a Stem-loop shape but the template
`DNA is not in a hairpin formation, the present hairpin PCR
`has the template DNA itself in a hairpin formation. This
`allows replication of both top and bottom strands of the
`template in a Single pass of the DNA polymerase and
`Subsequent conversion of polymerase errors to mismatches.
`0031 Gupte et al., U.S. Pat. Nos. 6,251,610; 6,258,544;
`and 6,087,099, describe the generation of a DNA hairpin
`during PCR, by joining top and bottom DNA strands, in
`order to allow DNA sequencing of both Strands in one pass.
`However, because their procedure requires polymerase
`extension (i.e. PCR) to generate the DNA strand-joining, it
`cannot be used to eliminate PCR errors since by the time the
`two Strands are joined together Some of the errors can have
`already occurred. (i.e. Since they start by a regular PCR
`reaction they have already lost the game in Step 1).
`0.032
`Oligonucleotide primers useful in the present
`invention can be Synthesized using established oligonucle
`otide Synthesis methods. Methods of Synthesizing oligo
`nucleotides are well known in the art. Such methods can
`range from Standard enzymatic digestion followed by nucle
`otide fragment isolation (see for example, Sambrook, et al.,
`
`Molecular Cloning: A Laboratory Manual, Second Edition,
`Cold Spring Harbor, N.Y., (1989), Wu et al, Methods in
`Gene Biotechnology (CRC Press, New York, N.Y., 1997),
`and Recombinant Gene Expression Protocols, in Methods in
`Molecular Biology, Vol. 62, (Tuan, ed., Humana Press,
`Totowa, N.J., 1997), the disclosures of which are hereby
`incorporated by reference) to purely Synthetic methods, for
`example, by the cyanoethyl phosphoramidite method using
`a Milligen or Beckman System 1 Plus DNA synthesizer (for
`example, Model 8700 automated synthesizer of Milligen
`Biosearch, Burlington, Mass. or ABI Model 380B). Syn
`thetic methods useful for making oligonucleotides are also
`described by Ikuta et al., Ann. Rev. Biochem. 53:323-356
`(1984), (phosphotriester and phosphite-triester methods),
`and Narang et al., Methods Enzymol., 65:610-620 (1980),
`(phosphotriester method). Protein nucleic acid molecules
`can be made using known methods Such as those described
`by Nielsen et al., Bioconjug. Chem. 5:3-7 (1994).
`0033 AS used herein, the term “primer' has the conven
`tional meaning associated with it in Standard nucleic acid
`procedures, i.e., an oligonucleotide that can hybridize to a
`polynucleotide template and act as a point of initiation for
`the Synthesis of a primer extension product that is comple
`mentary to the template Strand.
`0034. Many of the oligonucleotides described herein are
`designed to be complementary to certain portions of other
`oligonucleotides or nucleic acids Such that Stable hybrids
`can be formed between them. The Stability of these hybrids
`can be calculated using known methods Such as those
`described in Lesnick and Freier, Biochemistry 34:10807
`10815 (1995), McGraw et al., Biotechniques 8:674-678
`(1990), and Rychlik et al., Nucleic Acids Res. 18:6409-6412
`(1990).
`0035. The template nucleic acid that is to be amplified in
`a hairpin formation is preferably DNA, but it can also be
`RNA or a synthetic nucleic acid. The template can be of any
`size, but preferably of a size that can be replicated by DNA
`or RNA polymerases; most preferably the template in the
`region 50 bp-1000 base pairs.
`0036) The nucleic acid target can be any double stranded
`nucleic acid which is capable of being amplified.
`0037. The target nucleic acid can be from any source,
`Such as a PCR product of a known gene or a preparation of
`genomic DNA. The preferred target nucleic acid is DNA,
`but MRNA can also be used. The DNA can be any mixture
`containing one or various sizes of DNA, such as cDNA
`synthesized from the whole MRNA collected from cells that
`need to be Screened for mutation/polymorphism; or fractions
`thereof; or the whole genomic DNA collected from cells that
`need to be Screened for mutation/polymorphism; or fractions
`thereof; or any combination of the above digested into
`Smaller pieces by enzymes.
`0038 Any method of amplifying a nucleic acid target can
`be used. The amplification reaction can be polymerase chain
`reaction (PCR), ligase chain reaction (LCR), Strand dis
`placement amplification (SDA), transcription mediated
`amplification (TMA), O?3-replicase amplification (Q-beta),
`or rolling circle amplification (RCA).
`0039) Preferably, PCR is used to amplify the nucleic acid
`target.
`
`Oxford, Exh. 1006, p. 11
`
`

`

`US 2005/0142559 A1
`
`Jun. 30, 2005
`
`0040 Any polymerase which can synthesize the desired
`nucleic acid may be used. Preferred polymerases include but
`are not limited to Sequenase, Vent, and Taq polymerase.
`Preferably, one uses a high fidelity polymerase (Such as
`Clontech HF-2) to minimize polymerase-introduced muta
`tions.
`0041. In one preferred embodiment, rolling circle ampli
`fication (RCA) is used to amplify the nucleic acid template.
`Rolling circle amplification (RCA) is an isothermal process
`for generating multiple copies of a Sequence. In rolling circle
`DNA replication in vivo, a DNA polymerase extends a
`primer on a circular template (Komberg, A. and Baker, T. A.
`DNA Replication, W. H. Freeman, New York, 1991). The
`product consists of tandemly linked copies of the comple
`mentary Sequence of the template. RCA is a method that has
`been adapted for use in vitro for DNA amplification (Fire, A.
`and Si-Oun Xu, Proc. Natl. Acad Sci. USA, 1995, 92:4641
`4645; Lui, D., et al., J. Am. Chem. Soc., 1996, 118:1587
`1594; Lizardi, P. M., et al., Nature Genetics, 1998, 19:225
`232; U.S. Pat. No. 5,714,320 to Kool).
`0042. In RCA techniques a primer sequence having a
`region complementary to an amplification target circle
`(ATC) is combined with an ATC. Following hybridization,
`enzyme, dNTPs, etc. allow extension of the primer along the
`ATC template, with DNA polymerase displacing the earlier
`Segment, generating a Single Stranded DNA product which
`consists of repeated tandem units of the original ATC
`Sequence. RCA techniques are well known in the art, includ
`ing linear RCA (LRCA). Any such RCA technique can be
`used in the present invention.
`0043. When RCA is used to amplify the hairpin structure,
`Cap2 should not contain a polymerase block in order to
`allow the enzyme to continuously perform DNA synthesis
`on the circularized DNA template. In this approach, follow
`ing ligation of Cap 1 and Cap 2 a polymerase reaction is
`initiated by addition of a single primer that binds to the Cap
`2 non-complementary region. The polymerase then extends
`the primer by performing numerous circles around the
`original template, and resulting in a DNA amplification that
`copies both DNA strands every time it performs a full circle.
`Similar to non-isothermal amplification, during isothermal
`amplification too, every time there is a polymerase error
`during amplification it will form a mismatch while genuine
`changes such as mutations will be fully matched. Follow
`ing amplification, the original DNA sequence can be recov
`ered with a restriction digestion which separates the DNA
`caps introduced in the first Step of the procedure.
`0044) It is possible that instead of ligating Cap1 and Cap2
`to the template DNA for the purpose of generating a hairpin
`Structure with non complementary ends, the same result can
`be achieved via utilization of the first few steps described in
`t

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket