throbber
Digital quantification of mutant DNA in cancer patients
`Frank Diehla,b and Luis A. Diaz Jra
`
`Purpose of review
`The accumulation of somatic mutations is the major driving
`force for tumorigenesis. These mutations uniquely
`differentiate tumor cells from their normal counterparts.
`Mutations within tumor cells and mutant DNA released by
`tumor cells into blood, lymph, stool, tissues and other bodily
`compartments can thereby be used for cancer detection.
`Here we discuss technologies available for the detection
`and quantification of mutant DNA in clinical samples and the
`value of such measurements for patient management.
`Recent findings
`Conventional mutation detection technologies are either
`qualitative or only roughly estimate the abundance of mutant
`DNA molecules. Recently-developed approaches,
`however, use single molecule counting to determine the
`genotype of each individual member of a DNA population,
`providing a more accurate and precise digital output.
`Summary
`In this review, we discuss the clinical utility of mutant DNA
`quantification in cancer patients in the context of recent
`technical advances made in digital mutation detection.
`
`Keywords
`biomarker, molecular diagnostics, mutation, quantification,
`single molecule detection
`
`Curr Opin Oncol 19:36–42. ß 2007 Lippincott Williams & Wilkins.
`
`aThe Ludwig Center for Cancer Genetics and Therapeutics, Baltimore and bThe
`Howard Hughes Medical Institute, The Johns Hopkins Kimmel Cancer Center,
`Baltimore, Maryland, USA
`
`Correspondence to Frank Diehl, PhD, The Ludwig Center for Cancer Genetics and
`Therapeutics, The Johns Hopkins Kimmel Cancer Center, 1650 Orleans Street,
`Room 590, Baltimore, MD 21231, USA
`Tel: +1 410 955 8878; fax: +1 410 955 0548;
`e-mails: fdiehl2@jhmi.edu, frank_diehl@yahoo.com
`
`Conflict of Interest and Sponsorship Statements: No conflicts declared.
`
`Current Opinion in Oncology 2007, 19:36–42
`
`Abbreviations
`
`APC
`ATP
`BCR-ABL
`EBV
`dHPLC
`EGFR
`FRET
`PCR
`PSA
`RCA
`SBE
`SMD
`
`adenomatosis polyposis coli
`adenosine triphosphate
`breakpoint cluster region-Abelson
`Epstein-Barr virus
`denaturing high performance liquid chromatography
`epidermal growth factor receptor
`fluorescence resonance energy transfer
`polymerase chain reaction
`prostate specific antigen
`rolling-circle amplification
`single base extension
`single molecule detection
`
`ß 2007 Lippincott Williams & Wilkins
`1040-8746
`
`36
`
`Introduction
`The molecular basis of cancer is rapidly being deciphered
`and recent studies suggest that a complex array of genetic
`alterations exist in most human cancers [1]. Genetic
`alterations including gene deletions, gene amplifications,
`point mutations, and chromosomal rearrangements play a
`major role in the development and progression of cancers
`and are therefore unique identifiers that distinguish
`tumor cells from their normal counterparts.
`
`In order to best use gene alterations as a biomarker in
`clinical oncology, it must be possible to detect the tumor-
`specific genetic changes in a background of DNA from
`normal cells. Gene deletions and amplifications consist of
`copy number alterations,
`instead of changes in the
`primary DNA sequence and thus are not easily dis-
`tinguishable from DNA of normal cells. Detection and
`enumeration of gene copy number changes therefore
`require tumor cell isolation or direct cellular visualization.
`For example, in-situ hybridization or laser capture micro-
`dissection followed by real-time polymerase chain reac-
`tion (PCR) is used for the quantification of v-erb-b2
`erythroblastic leukemia viral oncogene homolog 2 ampli-
`fications in breast cancer tissue [2].
`
`Unlike deletions and amplifications, point mutations and
`chromosomal rearrangements represent changes of the
`primary DNA sequence that are substantially different
`from normal DNA and thus can be detected within
`clinical samples without the need for prior tumor cell
`isolation or visualization. Mutation detection has often
`been performed qualitatively without the ability to
`quantify the amount of mutant and wild-type DNA
`present in the sample. Qualitative mutation detection
`assays can have two potential problems. First, an assay
`could yield a false-negative result because the amount of
`starting DNA is too low to detect minority mutations.
`Second, an assay could yield a stochastic false-positive
`result because rare random mutations are present in a
`sample. Quantitative technologies could overcome these
`problems. They have the ability to directly measure the
`number of DNA molecules tested per assay and therefore
`ensure that the amount of starting material is sufficient to
`detect minority mutations in the predicted frequency
`range. Quantification also allows one to distinguish
`between random and pathogenic mutations by establish-
`ing a baseline mutant-to-wild-type ratio (e.g. the back-
`ground mutation frequency of human cells), where
`mutations found at a ratio below the baseline are con-
`sidered random. Quantitative assays also allow standard
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`The Johns Hopkins University Exhibit JHU2004 - Page 1 of 7
`
`

`

`quality control monitoring which is a necessary require-
`ment for routine diagnostic testing [3].
`
`Several quantitative mutation detection technologies are
`currently in development, and translational studies are
`needed to demonstrate whether quantification of mutant
`DNA is beneficial for early detection and as a biomarker
`of malignancy. Technologies attempting to quantify can-
`cer mutations and their potential clinical application are
`the focus of this review.
`
`Mutation detection and quantification
`principles
`Over the past decade, many techniques have been devel-
`oped for the analysis of mutated DNA. The impetus for
`this effort has been the desire to increase sensitivity,
`specificity and efficiency while decreasing cost. Analyti-
`cal sensitivity and specificity are parameters that refer to
`the lowest amount of mutant DNA that can be detected
`in a high background of normal DNA and the ability to
`exclusively detect actual mutations, respectively. Assay
`efficiency and cost are linked and refer to the time, labor
`and cost required for each analysis. The performance of
`the currently available technologies varies considerably.
`To better understand the variability, we grouped them
`based on (i) how many mutations can be analyzed per
`assay, (ii) the order of amplification and allele discrimi-
`nation, and (iii) the quantitative nature of the assay
`signals.
`
`Screening and locus specific mutation detection
`Screening or scanning methods can detect a range of
`possible mutations in a target region. They are particu-
`larly important for the discovery of new cancer genes [4].
`For this application, the sensitivity of standard Sanger
`sequencing is sufficient. The identification of minority
`mutations, however, requires higher sensitivities and
`specificities. Current scanning technologies are in general
`not sensitive enough, due to larger numbers of positions
`analyzed. Once a new mutation is associated with a
`certain cancer, it is usually sufficient to detect it by
`locus-specific methods. Locus-specific assays have the
`advantage of being more sensitive, cheaper and easier to
`perform. Several assays that can be used for the quanti-
`fication of known and unknown mutations are described
`below.
`
`Direct and indirect allele discrimination
`Most techniques rely on the following four components:
`(i) amplification of the target sequence, (ii) discrimination
`of the mutant and wild-type DNA sequences, (iii) sep-
`aration, and (iv) detection. Approaches that amplify the
`DNA before genotype discrimination are named indirect
`detection methods. The amplification is most commonly
`accomplished by the use of PCR and therefore suffers
`from typical PCR-associated complications, such as the
`
`Quantification of mutant DNA in cancer Diehl and Diaz 37
`
`generation of random errors by the DNA polymerase and
`allele bias that results in poor quantitative accuracy.
`Direct methods, on the other hand, discriminate the
`alleles prior to the amplification or detection step. The
`presence or absence of the mutation is determined by
`allele-specific methods. DNA ligases, DNA polymerases
`and DNA nucleases, as well as thermodynamic differ-
`ences between matched and mismatched DNA duplexes
`are used to discriminate between alleles. The initial
`allele discrimination step greatly influences the speci-
`ficity and sensitivity of any direct mutation detection test.
`For some assay platforms, the DNA molecules need to be
`separated before detection by binding to a solid phase or
`by gel electrophoresis. Unfortunately, the separation step
`introduces the risk of cross-contamination and requires
`additional handling of the samples. An assay format that
`avoids these problems is a homogeneous test that does
`not require separation and can combine the amplification,
`discrimination and detection step essentially in a single
`tube [5].
`
`Analog and digital quantification of mutations
`As discussed above, the assay strategy determines the
`specificity and sensitivity as well as the precision and
`accuracy of allele quantification. These parameters also
`depend on the nature of the assay signal generated.
`Traditional genotyping assays determine the identity
`of a particular base as an average contribution to a
`heterogeneous population of DNA molecules. Thus,
`such methods only convey an ‘analog’ signal for the
`individual members of the DNA pool. If more than
`one genotype is queried at a time, a ratio can be calcu-
`lated between the different alleles present in the reaction
`(Fig. 1a). Most assays available today are analog and have
`been reviewed elsewhere [5–7]. Here, we present
`examples of assays that have recently been used for
`quantification of mutations in clinical samples. Pyrose-
`quencing, for example, is a nucleotide extension sequen-
`cing approach where pyrophosphate is generated when a
`particular nucleotide anneals to the template and is
`incorporated by DNA polymerase. Subsequently, pyro-
`phosphate is converted to adenosine triphosphate (ATP)
`by ATP sulfurylase, which provides the energy for luci-
`ferase to oxidize luciferin and generate light. The inten-
`sity of the light is proportional to the amounts of annealed
`and extended nucleotide molecules. The peak sizes of
`the pyrogram are used to quantify the relative amount of
`each allele down to a mutant to wild-type ratio of 5% [8].
`Another example of analog mutation detection is a DNA
`endonuclease-based assay followed by denaturing high
`performance liquid chromatography (dHPLC). The
`quantitative output signals are the peak sizes on the
`dHPLC chromatograms. The assay can be used to
`quantify unknown mutations if present above 1% [9].
`Two final examples are the LigAmp assay and the Scor-
`pion primer-based quantitative PCR assay which can
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`The Johns Hopkins University Exhibit JHU2004 - Page 2 of 7
`
`

`

`38 Cancer biology
`
`Figure 1 Principle of analog and digital mutation analysis
`
`(a)
`
`(b)
`
`Analog
`
`Digital
`
`100%
`90%
`80%
`70%
`60%
`50%
`40%
`30%
`20%
`10%
`0%
`
`Wild-type
`
`Mutant
`
`100%
`90%
`80%
`70%
`60%
`50%
`40%
`30%
`20%
`10%
`0%
`
`1
`
`3
`
`5
`
`7
`
`9
`11
`13
`Wild-type
`
`17
`15
`Mutant
`
`19
`
`21
`
`23
`
`25
`
`(a) In analog assays, an average signal is acquired from the mutant and wild-type DNA molecules present in the sample. The ratio between the mutant
`and wild-type signal is an estimate of the mutation frequency. (b) In digital assays, the genotype of the individual DNA molecules is determined
`separately. Counting is used to quantify the mutant and wild-type DNA molecules present in the sample.
`
`quantify mutations down to 0.01% [10,11]. As homo-
`geneous assays, these measure in real-time the accumu-
`lation of mutant and wild-type PCR product during each
`cycle. In the log-linear phase of the amplification the
`amount of the target DNA correlates with the initial
`copy numbers.
`
`A more precise and accurate approach to mutation quanti-
`fication is based on discrete counting of the mutant and
`wild-type alleles present in a sample. Techniques incor-
`porating this methodology are termed ‘digital’ as they are
`able to generate binary results (mutation present or absent)
`on each individual member of a DNA pool (Fig. 1b)
`[12,13]. The quantitative precision and accuracy of
`digital assays is limited only by the number of molecules
`being analyzed. Statistics become important for counting
`rare events, which is the case when rare mutant alleles are
`present in a high background of wild-type molecules.
`Based on the Poisson distribution, the standard deviation
`of the number of rare events equals the square root of the
`
`number of detected events. Only the measured back-
`ground events need to be subtracted from the positives
`to get the net counts. This phenomenon has been called
`Poisson noise, and it limits the precision of single molecule
`detection methods [14]. The precision can be improved
`only by increasing the number of molecules analyzed. For
`example, a precision of 10% would require the detection of
`100 mutant molecules per measurement. Considering a
`sensitivity limit of 0.01% for the detection of mutant DNA
`in plasma, this would require the analysis of a total of
`1 106 DNA molecules. This translates into 3 mg of
`human genomic DNA, which exceeds the amount of
`DNA present in most plasma samples [15].
`
`Digital mutation quantification methods
`Various approaches have been described that allow the
`confined digital analysis of single molecules. We will
`review direct and indirect methods that can be used
`for mutation quantification in DNA fragments at known
`or unknown base positions (Table 1).
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`The Johns Hopkins University Exhibit JHU2004 - Page 3 of 7
`
`

`

`Quantification of mutant DNA in cancer Diehl and Diaz 39
`
`[40]
`[33,34]
`
`[38]
`[15]
`[36]
`
`[19]
`[23]
`[16]
`
`[22]
`
`References
`
`[29–32]
`
`K-ras
`
`1/100–1000
`
`p53
`
`1/100000a
`
`CFTR
`
`ND
`
`CFTR
`
`1/500–1500
`
`Flowcytometry
`platereader
`Fluorescence
`
`machine
`
`Real-timePCR
`microscope
`
`Scanningelectron
`
`Microscope
`Fluorescence
`
`EGFR
`
`1/1000
`
`GS20,454Corp.
`
`NA
`
`APC
`
`1/500
`
`Gelscanner
`
`PIK3CA,p53
`
`K-ras,
`
`1/10000
`
`Flowcytometry
`
`APC
`
`K-ras
`
`1/1000
`
`1/10000
`
`Flowcytometry
`
`PAGE
`
`separation
`
`Magnetic
`
`separation
`
`Magnetic
`
`separation
`
`Magnetic
`
`NA
`
`separation
`
`Magnetic
`
`NA
`
`NA
`
`NA
`
`K-ras
`
`Gene
`
`1/1000
`
`Microfluidicdevice
`
`Sensitivityb
`
`Detection
`
`Separation
`
`Direct digital mutation detection
`Direct digital quantification methods are based on the
`discrimination of the genotype of single DNA molecules
`followed by single molecule amplification and/or detec-
`tion. As described above, the enzymatic or physical
`methods used to distinguish mutant from wild-type
`sequences limit the sensitivity and specificity of these
`assays. For example, the initial allele discrimination of
`DNA molecules immobilized on glass slides can be done
`using a ligation assay [16] or a surface-invasion cleavage
`assay [17]. These strategies can at best detect one mutant
`in 1500 wild-type molecules. After the allele discrimi-
`nation reaction,
`allele-specific
`circularized single-
`stranded DNA molecules are hybridized to the ligation
`or cleavage products and used as a template for sub-
`sequent rolling-circle amplification (RCA). RCA pro-
`duces a long single-stranded concatemerized molecule
`containing multiple copies of the complementary circular
`starting sequence. The amplified DNA sequences are
`then labeled by hybridizing allele-specific fluorescent
`oligonucleotides to the RCA products. The image of
`the slide taken after the labeling is used to count the
`individual molecules. Recently, a similar solution-based
`approach was used for the digital analysis of DNA from
`pathogens [18]. This homogeneous test has the poten-
`tial to be adapted to mutation detection and quantifi-
`cation. Briefly, the assay uses ligation for the circulariza-
`tion of padlock probes,
`subsequent RCA, and a
`microfluidic device attached to a fluorescence microscope
`to count the molecules. Another type of assay allows the
`absolute quantification of unknown mutations in DNA
`fragments, but not the number of wild-type DNA mol-
`ecules [19]. The test is based on capturing the target
`sequences on beads coated with complementary probes,
`selective digestion of wild-type molecules, and the sub-
`sequent quantification of intact mutant molecules by
`digital PCR (see below). The high sensitivity of this test
`(1/100 000) can only be accomplished by performing
`several rounds of mutant enrichment, which unfortu-
`nately makes the assay difficult to use for clinical appli-
`cations.
`
`Several years ago, fluorescence-based single molecule
`detection (SMD) approaches were introduced. These
`are based on the direct visualization of individual fluor-
`escently labeled DNA molecules without the need for
`enzymatic amplification [20,21] So far only one of these
`approaches has been used for the enumeration of point
`mutations in solution [22]. The assay is based on the
`allele-specific ligation of fluorescence resonance energy
`transfer (FRET) probes generating molecular beacons
`upon successful ligation. A laser-based fluorescence sys-
`tem attached to a heatable microfluidic device is used to
`detect photon bursts generated by the FRET reaction.
`SMD technologies have also been developed for the
`analysis of single DNA molecules immobilized on solid
`
`Pyrosequencing
`
`nonmagneticbeads
`
`Emulsion,PCRon
`
`Proteintruncationtest
`
`Singlebaseextension
`
`Singlebaseextension
`
`Molecularbeacons
`
`PCRinsolution
`
`magneticbeads
`Emulsion,PCRon
`magneticbeads
`Emulsion,PCRon
`magneticbeads
`Emulsion,PCRon
`
`bMutant/wild-type.
`aQuantificationofmutantpopulationonly;
`NA,notapplicable;ND,notdetermined.
`
`Indirect,scanning
`
`stopcodons
`
`Indirect,scanningfor
`
`Indirect,locus-specific
`
`Indirect,locus-specific
`
`pyrosequencing
`
`DigitalPCRand
`truncationtest
`
`DigitalPCRandprotein
`singlebaseextension
`
`DigitalPCR,RCAand
`
`baseextension
`
`DigitalPCRandsingle
`beaconhybridization
`
`Indirect,locus-specific
`
`DigitalPCRandmolecular
`
`beaconhybridization
`
`Molecularbeacons
`
`PCRinsolution
`
`Indirect,locus-specific
`
`DigitalPCRandmolecular
`
`Restrictiondigest
`
`PCRinsolution
`
`Direct,locus-specific
`
`Invasivecleave
`
`oligonucleotides
`
`Ligationof
`
`molecularbeacons
`
`Ligationof
`
`NA
`
`Direct,locus-specific
`
`RCA
`
`Direct,locus-specific
`
`LigationandRCA
`
`NA
`
`Direct,locus-specific
`
`LigationandFRET
`
`digitalPCR
`
`Restrictiondigestand
`
`goldnanoparticles
`Invasivecleavageand
`
`Discrimination
`
`Amplification
`
`Assayprinciple
`
`Technique
`
`Table1Digitalassaysappliedtomutationdetection
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`The Johns Hopkins University Exhibit JHU2004 - Page 4 of 7
`
`

`

`40 Cancer biology
`
`there is only one base-specific
`supports. Currently,
`approach described in the literature that could readily
`be applied to the enumeration of mutations. The test is
`based on a surface-invasion cleavage assay followed by
`the detection of mutant alleles using gold nanoparticles
`[23]. Recently, technologies for the sequencing of single
`DNA molecules on solid support have also been intro-
`duced. These are intended for the low-cost resequencing
`of complete genomes, but could eventually be adapted
`for the quantification of genetic variations in clinical
`samples [24–26]. Most notable are two approaches that
`use cyclic array sequencing. These rely on the extension
`of a DNA template hybridized to immobilized primers by
`a polymerase in the presence of fluorescently-labeled
`nucleotides [25,27]. Unfortunately, the sensitivity of
`these methods is limited by the error rates of the
`DNA polymerases currently available for sequencing.
`These error rates are at least one log higher compared
`with DNA polymerases conventionally used for PCR.
`
`Indirect digital mutation detection
`Indirect digital mutation quantification involves an
`initial compartmentalized amplification of single DNA
`molecules followed by allele discrimination and detec-
`tion. Cloning of DNA fragments followed by the sequen-
`cing of individual bacterial colonies is the most basic
`form of digital analysis. Unfortunately, this approach is
`time-consuming and labor-intensive. In 1988, it was
`demonstrated that PCR can be efficiently performed
`on single DNA templates [28]. This opened the way
`for several applications, reviewed elsewhere [12,13].
`One such technique, which came to be known as digital
`PCR, proved to be a powerful tool for single molecule
`counting and quantification of somatic mutations in
`clinical samples [29]. In digital PCR assays, multiple
`PCR reactions are performed in parallel at DNA con-
`centrations so low that most reactions contain zero or one
`template molecule and thus can be amplified clonally.
`Each resulting DNA pool is then analyzed individually
`for the presence of mutant and wild-type sequences by
`using fluorescent allele-specific molecular beacons. The
`digital PCR approach has been used for detection of
`K-ras mutations in various clinical samples [29 –32].
`Protein truncation tests which can be used to scan for
`stop codons within a target sequence have also been
`combined with digital PCR [33,34]. In contrast to more
`conventional mutation detection methods, the sensi-
`tivity of digital PCR approaches is not limited by the
`DNA polymerase error rate. In the worst-case scenario,
`an error occurs in the first cycle of a double-stranded
`single molecule PCR reaction which will result in a
`mutant to wild-type ratio of one to four. This would
`translate to a mixed mutant/wild-type signal
`in the
`subsequent genotyping assay and thus be excluded from
`the calculation. The sensitivity is instead limited by the
`number of molecules that can be analyzed and the false-
`
`positive rate of the mutation detection assay. In prin-
`ciple, the latter has the least influence on the detection
`limit as the analysis only needs to distinguish analytes
`that are exclusively wild-type or exclusively mutant.
`The main limitation of the originally described digital
`PCR techniques was the cost and labor involved in
`performing a large number of individual PCR reactions.
`To address these issues, several methods allowing
`millions of single-molecule PCR reactions to be per-
`formed in a single assay have been developed. One way
`to achieve this is by performing single-molecule PCRs in
`a thin polyacrylamide film poured on a glass microscope
`slide [35]. The amplification results in discrete DNA
`colonies in the polymer matrix (polonies). Another
`approach involves BEAMing (beads, emulsions, ampli-
`fication and magnetics) which allows single-molecule
`PCR reactions to be performed on magnetic beads in
`water-in-oil emulsions [36]. An alternative way to gen-
`erate beads coated with clonally amplified PCR products
`is by adding single beads into wells of a picotiter plate
`that can be used for digital PCR [37]. The bead suspen-
`sion obtained after solid-phase PCR accurately reflects
`the DNA diversity present in the template population
`and can therefore be used for mutation quantification.
`Recently, BEAMing followed by single base extension
`(SBE) and flow cytometry was used to quantify the level
`of mutated DNA circulating in the plasma of colorectal
`]. The detection limit of this assay
`tumor patients [15
`was one mutant DNA molecule in a background of 10 000
`wild-type DNA molecules. This threshold was deter-
`mined by the error rate of the DNA polymerase used for
`the preamplification of the limited amounts of plasma
`DNA. The sensitivity of BEAMing-based assays can also
`be constrained by the signal-to-noise ratio of the bead-
`based SBE assay. Therefore, RCA was used to increase
`the DNA copy number on the beads, making the SBE
`assay more specific [38]. Besides analyzing a mutation at
`a specific location, the DNA on beads can also be used as
`a template for sequencing. 454 Life Sciences developed
`an approach to sequence DNA on individual beads in
`parallel [39]. This system is commercially available and
`has been used for the identification and quantification of
`epidermal growth factor receptor (EGFR) gene mutations in
`lung cancer biopsies [40]. Another strategy used DNA
`coated beads immobilized in a polyacrylamide gel as a
`template for cycle sequencing by ligation [41].
`
`The clinical application of counting mutations
`Cancers are currently managed by a variety of clinical
`markers, which generally include patient symptoms,
`radiographic evaluation,
`routine laboratory tests and
`pathologic evaluation. These markers are used not only
`for diagnosis, but also as prognostic and predictive mar-
`kers, for tumor staging, and as markers for tumor response
`and detection of residual disease. More sensitive and
`specific biomarkers could aid cancer diagnosis and man-
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`The Johns Hopkins University Exhibit JHU2004 - Page 5 of 7
`
`

`

`Quantification of mutant DNA in cancer Diehl and Diaz 41
`
`Conclusions
`The development of digital technologies has resulted in
`the ability to precisely and accurately count mutant and
`normal DNA molecules present in a sample. This is an
`improvement over the analog approaches, which can only
`estimate relative amounts. Indirect digital PCR-based
`assays are also more sensitive. When digital assays are
`performed in a highly parallel setup that only requires low
`amounts of enzymes, digital and analog approaches run at
`a similar cost.
`
`Beyond the application of early detection and mutation
`defining diagnosis, mutant DNA quantification could also
`be used to estimate tumor burden in already diagnosed
`cancer patients. The amount of mutant DNA could
`predict recurrence more specifically than currently avail-
`able protein biomarkers. We envision a simple mutation-
`specific blood test that can diagnose cancer at earlier
`stages than before and measure patients’ tumor burden.
`In addition, a blood test of this nature will have the
`potential to define therapy based on tumor-specific
`mutations and/or detect treatment-resistant mutations
`as they arise.
`
`Acknowledgements
`We thank Will Hendricks, Nickolas Papadopoulos, Ian Cheong, Kerstin
`Schmidt, and Bert Vogelstein for critical review of the manuscript.
`
`References and recommended reading
`Papers of particular interest, published within the annual period of review, have
`been highlighted as:
`
`of special interest
` of outstanding interest
`Additional references related to this topic can also be found in the Current
`World Literature section in this issue (pp. 71–74).
`
`1
`
`2
`
`Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat
`Med 2004; 10:789–799.
`
`Tokunaga E, Oki E, Nishida K, et al. Trastuzumab and breast cancer: devel-
`opments and current status. Int J Clin Oncol 2006; 11:199–208.
`
`3
`
`Strom CM. Mutation detection, interpretation, and applications in the clinical
`laboratory setting. Mutat Res 2005; 573 (1–2):160–167.
`This review highlights the role of quantitative assays in diagnostic testing.
`
`4
`
`5
`
`Sjoblom T, Jones S, Wood LD, et al. The consensus coding sequences of
`human breast and colorectal cancers. Science 2006 [Epub ahead of print].
`
`Foy CA, Parkes HC. Emerging homogeneous DNA-based technologies in the
`clinical laboratory. Clin Chem 2001; 47:990–1000.
`
`6 Whitcombe D, Newton CR, Little S. Advances in approaches to DNA-based
`diagnostics. Curr Opin Biotechnol 1998; 9:602–608.
`
`7 Mike Makrigiorgos G. PCR-based detection of minority point mutations. Hum
`Mutat 2004; 23:406–412.
`
`8 Ogino S, Kawasaki T, Brahmandam M, et al. Sensitive sequencing method for
`KRAS mutation detection by pyrosequencing. J Mol Diagn 2005; 7:413–
`421.
`
`9
`
`Janne PA, Borras AM, Kuang Y, et al. A rapid and sensitive enzymatic method
`for epidermal growth factor receptor mutation screening. Clin Cancer Res
`2006; 12 (3 Pt 1):751–758.
`
`10 Shi C, Eshleman SH, Jones D, et al. LigAmp for sensitive detection of single-
`nucleotide differences. Nat Methods 2004; 1:141–147.
`
`11
`
` Kimura H, Kasahara K, Kawaishi M, et al. Detection of epidermal growth factor
`
`receptor mutations in serum as a predictor of the response to gefitinib in
`patients with nonsmall-cell lung cancer. Clin Cancer Res 2006; 12:3915–
`3921.
`Report on a blood-based assay to detect EGFR mutations in lung cancer. They
`used a Scorpion Amplified Refractory Mutation System for mutation detection.
`
`agement by giving physicians additional information to
`make decisions. For example, PSA is a widely-used
`marker available for prostate cancer, but lacks specificity
`to be used independently for the diagnosis of malignancy
`[42]. Markers for other solid cancers currently used in the
`clinic suffer from similar problems.
`
`New markers that are currently being developed are
`either based on the in-vivo or ex-vivo detection of cancer.
`In-vivo detection strategies rely on imaging technologies
`and have been reviewed elsewhere [43]. Biomarkers for
`ex-vivo detection most commonly include cancer-specific
`proteins or nucleic acids that can be assayed within cancer
`tissues, blood or other bodily fluids collected from cancer
`patients [44,45]. Of these markers, gene mutations are
`arguably the most specific. Thus far, however, mutations
`have only been routinely applied in the clinic for the
`diagnosis of hereditary cancer syndromes. To apply
`mutation detection for early detection and management
`of malignancies, sensitive and specific assays are essen-
`tial. Several investigational studies have started using
`quantitative approaches for this purpose. These studies
`include the noninvasive detection of mutant adenomatosis
`polyposis coli (APC) DNA in stool or plasma from subjects
`with colorectal
`adenomatous polyps
`and cancers
`[15,33]. Furthermore, the detection of mutations in
`the EGFR gene in biopsies and plasma has been under
`investigation partly due to a number of reports showing
`that these mutations are directly related to the efficacy of
`treatments targeting EGFR [9,11,40].
`
`Quantitative diagnostics have also become an essential
`part of the management of chronic myeloid leukemia
`patients treated with imatinib mesylate (Gleevec), a drug
`that appears to target the breakpoint cluster region Abel-
`son (BCR-ABL) fusion protein [46]. A real-time quanti-
`tative reverse transcriptase PCR (RT-PCR) assay of the
`BCR-ABL RNA transcript provides a measure of the total
`leukemia cell mass and thus can be used for the monitor-
`ing of patients. In fact, the rate and size of change in
`BCR-ABL load can predict long-term response to therapy
`and potential cure. The remaining challenge will be the
`standardization of BCR-ABL quantification across differ-
`ent institutions [47]. Another approach that may become
`standard in clinical practice is the quantification of
`Epstein-Barr virus (EBV) DNA in plasma, which is
`closely associated with nasopharyngeal carcinoma [48].
`It appears that plasma EBV DNA is useful for determin-
`ing prognosis and monitoring response to treatment. New
`concepts that could be applied to a broader range of
`tumors, in particular solid malignancies, are currently
`being investigated. These are based on the counting of
`intact tumor cells in the bone marrow or circulation
`[49,50], the quantification of DNA with tumor-specific
`methylation patterns [51], or the detection of tumor-
`specific RNA [52].
`
`Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
`
`The Johns Hopkins University Exhibit JHU2004 - Page 6 of 7
`
`

`

`42 Cancer biology
`
`12 Pohl G, Shih Ie M. Principle and applications of digital PCR. Expert Rev Mol
`Diagn 2004; 4:41–47.
`
`33 Traverso G, Shuber A, Levin B, et al. Detection of APC mutations in fecal DNA
`from patients with colorectal tumors. N Engl J Med 2002; 346:311–320.
`
`13
`
` Kraytsberg Y, Khrapko K. Single-molecule PCR: an artifact-free PCR
`
`approach for the analysis of somatic mutations. Expert Rev Mol Diagn
`2005; 5:809–815.
`Recent review about digital PCR approaches for the analysis of somatic mutations.
`
`14 Castro A, Williams JG. Single-molecule detection of specific nucleic acid
`sequences in unamplified genomic DNA. Anal Chem 1997; 69:3915–
`3920.
`
`15 Diehl F, Li M, Dressman D, et al. Detection and quantification of mutations in
`
`the plasma of patients with colorectal tumors. Proc Natl Acad Sci USA 2005;
`102:16368–16373.
`Application of BEAMing for the quantification of mutant DNA in plasma from
`cancer patients.
`
`16 Lizardi PM, Huang X, Zhu Z, et al. Mutation detection and single-molecule
`counting using isothermal
`rolling-circle amplification. Nat Genet 1998;
`19:225–232.
`
`17 Nie B, Shortreed MR, Smith LM. Scoring single-nucleotide polymorphisms at
`the single-molecule level by counting individual DNA cleavage events on
`surfaces. Anal Chem 2005; 77:6594–6600.
`
`18
`Jarvius J, Melin J, Goransson J, et al. Digital quantification using amplified
`
`single-molecule detection. Nat Methods 2006; 3:725–727.

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